chromosome abnormalities and genetic counseling
OXFORD MONOGRAPHS ON MEDICAL GENETICS General Editors: judith g. hall
peter s. harper
louanne hudgkins
evan eichler
charles j. epstein†*
arno g. motulsky
Former Editors: charles scriver
martin bobrow
j.a. fraser roberts
c.o. carter
han brenner
1. R.B. McConnell: The genetics of gastrointestinal disorders 2. A.C. Kopéc: The distribution of the blood groups in the United Kingdom 3. E. Slater and V.A. Cowie: The genetics of mental disorders 4. C.O. Carter and T.J. Fairbank: the genetics of locomotor disorders 5. A.E. Mourant, A.C. Kopéc, and K. Domaniewska-Sobezak: The distribution of the human blood groups and other polymorphisms 6. A.E. Mourant, A.C. Kopéc, and K. Domaniewska-Sobezak: Blood groups and diseases 7. A.G. Steinbert and C.E. Cook: The distribution of the human immunoglobulin allotypes 8. D. Tills, A.C. Kopéc, and R.E. Tills The distribution of the human blood groups and other polymorphisms: Supplement I 9. D.Z. Loesch: Quantitative dermatoglyphics: classification, genetics, and pathology 10. D.J. Bond and A.C. Chandley: Aneuploidy 11. P.F. Benson and A.H. Fensom: Genetic biochemical disorders 12. G.R. Sutherland and F. Hecht: Fragile sites on human chromosomes 13. M. d’A Crawfurd: The genetics of renal tract disorders 14. C.R. Scriver and B. Child: Garrod’s inborn factors in disease 18. M. Baraitser: The genetics of neurological disorders 19. R.J. Gorlin, M.M. Cohen, Jr. and L.S. Levin: Syndromes of the head and neck, third edition 21. D. Warburton, J. Byrne, and N. Canki: Chromosome anomalies and prenatal development: an atlas 22. J.J. Nora, K. Berg, and A.H. Nora: Cardiovascular disease: genetics, epidemiology, and prevention 24. A.E.H. Emery: Duchenne muscular dystrophy, second edition 25. E.G.D. Tuddenham and D.N. Cooper: The molecular genetics of haemostasis and its inherited disorders 26. A. Boué: Foetal medicine 27. R.E. Stevenson, J.G. Hall, and R.M. Goodman: Human malformations 28. R.J. Gorlin, H.V. Toriello, and M.M. Cohen, Jr.: Hereditary hearing loss and its syndromes 29. R.J. M. Gardner and G. R. Sutherland: Chromosomes abnormalities and genetic counseling, second edition 30. A.S. Teebi and T.I. Farag: Genetic disorders among Arab populations 31. M.M. Cohen, Jr.: The child with multiple birth defects 32. W.W. Weber: Pharmacogenetics 33. V.P. Sybert: Genetic skin disorders 34. M. Baraitser: Genetics of neurological disorders, third edition 35. H. Ostrer: Non-Mendelian genetics in humans 36. E. Traboulsi: Genetic factors in human disease 37. G.L. Semenza: Transcription factors and human disease 38. L. Pinsky, R.P. Erickson, and R.N. Schimke: Genetic disorders of human sexual development 39. R.E. Stevenson, C.E. Schwartz, and R. J. Schroer: X-linked mental retardation 40. M.J. Khoury, W. Burke, and E. & Thomson: Genetics and public health in the 21st century 41. J. Weil: Psychosocial genetic counseling 42. R.J. Gorlin, M.M. Cohen, Jr., and R.C.M. Hennekam: Syndromes of the head and neck, fourth edition 43. M.M. Cohen, Jr., G. Neri, and R. Weksberg: Overgrowth syndromes 44. R.A. King, J.I. Rotter, and A.G. Motulsky: & e genetic basis of common diseases, second edition 45. G.P. Bates, P.S. Harper, and L. Jones: Huntington’s disease, third edition 46. R.J.M. Gardner and G.R. Sutherland: Chromosome abnormalities and genetic counseling, third edition 47. I.J. Holt: Genetics of mitochondrial disease 48. F. Flinter, E. Maher, and A. Saggar-Malik: & e genetics of renal disease 49. C.J. Epstein, R.P. Erickson, and A. Wynshaw-Boris: Inborn errors of development: the molecular basis of clinical disorders of morphogenesis 50. H.V. Toriello, W. Reardon, and R.J. Gorlin: Hereditary hearing loss and its syndromes, second edition 51. P. S. Harper: Landmarks in medical genetics 52. R.E. Stevenson and J.G. Hall: Human malformations and related anomalies, second edition 53. D. Kumar and S.D. Weatherall: Genomics and clinical medicine 54. C.J. Epstein, R.P. Erickson, and A. Wynshaw-Boris: Inborn errors of development: the molecular basis of clinical disorders of morphogenesis, second edition 55. W. Weber: Pharmacogenetics, second edition 56. P.L. Beales, I.S. Farooqi, and S. O’Rahilly: The genetics of obesity syndromes 57. P.S. Harper: A short history of medical genetics 58. R.C.M. Hennekam, I.D. Krantz, and J.E. Allanson: Gorlin’s syndromes of the head and neck, fifth edition 59. D. Kumar and P. Elliot: Principles and practices of cardiovascular genetics 60. V.P. Sybert: Genetic skin disorders, second edition 61. R.J.M. Gardner, G.R. Sutherland, and L.G. Shaffer: Chromosome Abnormalities and Genetic Counseling, fourth edition
†
Deceased
oxford monographs on medical genetics no. 61
chromosome abnormalities and genetic counseling FOURTH EDITI O N
R. J. McKinlay GARDNER ADJUNCT PROFESSOR U N I V E R S I T Y O F OTA G O , D U N E D I N , N E W Z E A L A N D AND NORTHERN REGIONAL GENETIC SERVICES AUCKLAND, NEW ZEALAND ( L AT E LY O F G E N E T I C H E A LT H S E R V I C E S V I C T O R I A MELBOURNE, AUSTRALIA)
Grant R. SUTHERLAND EMERITUS GENETICIST W O M E N ’ S A N D C H I L D R E N ’ S H O S P I TA L , A N D A F F I L I AT E P R O F E S S O R UNIVERSITY OF ADELAIDE, ADELAIDE, AUSTRALIA
Lisa G. SHAFFER S I G N AT U R E G E N O M I C L A B O R AT O R I E S S P O K A N E , W A S H I N G T O N , U N I T E D S TAT E S O F A M E R I C A
1
1 Oxford University Press, Inc., publishes works that further Oxford University’s objective of excellence in research, scholarship, and education. Oxford New York Auckland Cape Town Dar es Salaam Hong Kong Karachi Kuala Lumpur Madrid Melbourne Mexico City Nairobi New Delhi Shanghai Taipei Toronto With offices in Argentina Austria Brazil Chile Czech Republic France Greece Guatemala Hungary Italy Japan Poland Portugal Singapore South Korea Switzerland Thailand Turkey Ukraine Vietnam
Copyright © 2012 by Oxford University Press, Inc. Published by Oxford University Press, Inc. 198 Madison Avenue, New York, New York 10016 www.oup.com Oxford is a registered trademark of Oxford University Press All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, electronic, mechanical, photocopying, recording, or otherwise, without the prior permission of Oxford University Press. ____________________________________________________________________________________ Library of Congress Cataloging-in-Publication Data Gardner, R. J. M. Chromosome abnormalities and genetic counseling / R.J. McKinlay Gardner, Grant R. Sutherland, Lisa G. Shaffer. — 4th ed. p. ; cm. — (Oxford monographs on medical genetics ; no. 61) Includes bibliographical references and index. ISBN 978-0-19-537533-6 1. Genetic counseling. 2. Human chromosome abnormalities—Patients—Counseling of. I. Sutherland, Grant R. II. Shaffer, Lisa G. III. Title. IV. Series: Oxford monographs on medical genetics ; no. 61. [DNLM: 1. Chromosome Aberrations. 2. Genetic Counseling. QS 677] RB155.7.G37 2011 616’.042—dc22 2011005028 ____________________________________________________________________________________
987654321 Printed in USA on acid-free paper
This book is dedicated to Jocelyn, Geoffrey, and Craig, their parents, and all other families who seek our “chromosomal advice.”
Jocelyn and Geoffrey (with lamb) have a partial trisomy for chromosome 4 long arm, and Craig, the youngest, had a 46,XY result on amniocentesis. Their father is a translocation carrier (see Fig. 5–1, lower).
Heredity Inescapably, this is me—the diagnosis is cause for anger at those who brightly say we choose our destinies. There is no store of courage, wit or will can save me from myself and I must face my children, feeling like that wicked fairy, uninvited at the christening, bestowing on my own, amidst murmurs of apprehension, a most unwanted gift—that of a blighted mind. No one could tell me of this curse when I was young and dreamt of children and the graces they would bear. Later, it seemed that a chill morning revealed deeper layers of truth. For my romancing there is a price to pay— perhaps my children’s children will pass this tollgate after me. My grandmothers gaze down from their frames on my wall, sadly wondering. —Meg Campbell
Where is the wisdom we have lost in knowledge? Where is the knowledge we have lost in information? —T. S. Eliot Curiosity is a virtue, perhaps an unsung and undervalued virtue, which should be the energizing fuel to the thinking geneticist. —Willie Reardon
preface to fourth edition
Plus ça change, plus c’est la même chose. Classicists see the chromosome as a vertical structure; to the molecular scientist, DNA may lie horizontally: but they are looking at the same thing. The move away from classical microscopy into molecular methodologies, predicted for some time, is now actually happening. Smaller and subtler abnormalities can be detected. We know a lot more (but much yet to know) about normal genomic variation. Chromosomal testing of embryos before implantation, while not commonplace, is no longer regarded as extraordinary. Much progress is being made in enabling screening in pregnancy to be done earlier, and with greater precision. Be all this as it may, human biology remains human biology. Meiosis is as vulnerable as it always has been. Chromosomes continue to undergo rearrangement. Conceptions still happen, and children
continue to be born, with an incorrect chromosomal complement. And people still want to know why, and what they can do about it. The distinction between chromosome abnormalities and mutations in single gene diseases has become somewhat blurred. Some single gene mutations can be due to relatively large deletions, and some “cytogenetically” detected abnormalities can be deletions and duplications of smaller size. We deal primarily with those conditions that are detected in the expanded molecular cytogenetics laboratory, being aware that, for some of these, the risk assessments will follow those of dominant disorders. We have retained the fragile X syndromes, since these originated as chromosome disorders; in some respects, the molecular approach has simplified counselling, but in other ways, it has become more complicated. A twenty-first-century chromosome
•
vii
report is a sophisticated document, and it behoves the counselor to read it with an informed mind. We have therefore added a chapter devoted to the new technologies. So this book is changing, but it is also the same thing. We can answer some questions more accurately, indeed a lot more accurately, than hitherto;
viii •
P R E FA C E TO F O U RT H E D I T I O N
but mostly, these are questions we’ve heard many times before. Dunedin and Auckland Adelaide Spokane
R.J.M.G. G.R.S. L.G.S.
preface to the first edition
We have written this book with two particular categories of reader in mind. The first, and chief, audience is the genetic counselor, the person whose role it is to explain and interpret a chromosomal problem to the individual or family in whom it has been identified. Second, we have considered the needs of cytogenetic laboratory workers, who in addition to possessing the technical skills for chromosomal analysis, should understand the theoretical basis of clinical cytogenetics and be aware of the practical implications of their work. A pithy definition of counseling is assisting clients to recognize the nature of their problems and to find their own solutions to them. The description applies aptly to the specific case of genetic counseling. One tool the counselor needs to do the job well is accurate information about the nature of the problem. The aim of this book is to provide in convenient form accurate information concerning chromosomal conditions. We address these major questions clients have: How and why did it happen? Could it happen again? If so, how likely would it be to happen
again? Most chapters follow the general format of a Biology section, which discusses the first question, and a Genetic Counseling section, which deals with the latter two. (The division is not always clear-cut, and recognizing that readers may at times wish to refer just to the Genetic Counseling section, there is some duplication.) We do not claim, in using the expression “genetic counseling,” to encompass all that is involved in this multifaceted process. Rather, we seek to provide information and figures that will assist the client to decide what, for them, would be an appropriate course of action and to provide pointers to assist the counselor in communicating that information. Chromosomes are not a theoretical concept; they are real things. (In this spirit, it is helpful in counseling sessions to have on hand a real karyotype, preferably from the counselee’s own family, for use as a teaching aid.) To be able to give to the concept of chromosomes being passed down from parent to child a sense of reality, the counselor should have a broad and comfortable understanding •
ix
of the mechanisms of chromosome behavior. Thus, we have gone into considerable detail in describing particular chromosomal situations. Rare conditions are given almost as extensive a hearing as the more common abnormalities. We make these points: First, rare conditions, for the families that have them, are very common. Second, the intellectual exercise of coming to grips with the complexities of a variety of disorders sharpens the reader’s thinking. Is there a risk of the same (or another) chromosomal defect occurring in a future pregnancy, and leading to the birth of an abnormal child? And if such a risk exists, how great is it? The perception of risk is subjective. The fact that in this book we give probabilities to perhaps a fraction of one percent does not allow the counselor the comfort of supposing that interpreting risk to consultands is a neat and tidy business (Pearn, 1977; Bloch et al., 1979; Wertz et al., 1986). Sir William Osler said of clinical medicine that what kind of disease the patient had was not as important as what kind of patient had the disease. In a similar vein, we may remark that what kind of person faces a genetic risk warrants no less consideration than the degree of risk. But of course, a precise estimate does provide a major reference point to assist in making the subsequent decision. It is good to have good figures. Dr. Chapman explores the idea of “good figures” in Chapter 3. Other than peripherally, we do not describe details of karyotype–phenotype correlation (except in Chapter 22, where we directly address the problem of interpreting and managing unexpected chromosome abnormalities discovered in the course of prenatal diagnosis). This aspect of clinical cytogenetics is dealt with well in other texts. We will presume the reader has access to these and will not rework the information they contain. Likewise, we take it for granted the reader knows that skill in sensitive communication is an absolute requirement for effective genetic counseling. It is a continuing challenge to be able to communicate the gist of a situation in straightforward language, at the
x
•
P R E FA C E TO T H E F I R S T E D I T I O N
client’s level of comprehension. We recognize that however well done the cytogenetics and the risk determination, if counselees are not able to absorb this information and to react to it in a way that is most consonant with their own values, aspirations, and interests, the exercise has been less than successful. We do not explore the psychodynamics of counseling or the processes by which people make decisions: these issues are well addressed in other sources (Kelly, 1977; Hsia et al., 1979; Kessler, 1979; Epstein et al., 1979; Reed, 1980; Emery and Pullen, 1984; Reif and Baitsch, 1985; Hsia, 1987). A course couples need to consider when an abnormality is shown at prenatal diagnosis is termination of the pregnancy (“genetic abortion”). We assume the counselor is well aware of the support needed through this most difficult time (Landenburger and Delp, 1987; Magyari et al., 1987; Thomson, 1987). We have, more than once, taken some liberties of interpretation, and made some statements a little more firmly and less complicatedly than the current state of cytogenetic knowledge might allow. We trust that this will not undermine the validity of the book, bearing in mind its intended purpose—to be of use to both the experienced and the neophyte. “Families pursue genetic counseling in an effort to demystify the mysterious. They seek answers and information. If they did not want to ‘hear it all,’ they would not bother with genetic counseling. Families want an honest evaluation of what is known and what is unknown, a clear explanation of all possibilities, both good and bad, and a sensitive exploration of all available information with which they can make knowledgeable decisions about future family planning.” Thus Bloch et al. (1979) succinctly convey the essence of why people go to the genetic counselor. We hope this book will assist counselors in their task. January Dunedin, New Zealand Adelaide, Australia
1989 R.J.M.G. G.R.S.
acknowledgments
We thank the following for their expert advice and assistance in particular chapters: David Amor, Elizabeth Baker, John Barber, Kathie Friend, Alice George, Randi Hagerman, Jane Halliday, Brynn Levy, Donald Love, Alina Midro, Stephen Robertson, Trilochan Sahoo, Dianne Webster, and Leeanda Wilton. Aaron Theisen and Erin Dodge gave sterling
service in organizing the references. Kelley Gardner redrew some figures, and provided invaluable IT support. We acknowledge the National Institutes of Health, whose PubMed facility has, in large part, enabled the literature tracking that underpins the material in this book.
•
xi
This page intentionally left blank
contents
PART ONE: BASIC CONCEPTS
1. Elements of Medical Cytogenetics 3 2. Chromosome Analysis 21 3. The Origins and Consequences of Chromosome Pathology 27 4. Deriving and Using a Risk Figure 59
8. Centromere Fissions, Complementary Isochromosomes, Telomeric Fusions, Balancing Supernumerary Chromosomes, and Jumping Translocations 155 9. Inversions 161 10. Insertions 183 11. Autosomal Ring Chromosomes 201
PART TWO: PARENT WITH A CHROMOSOMAL ABNORMALITY
5. Autosomal Reciprocal Translocations 67 6. Sex Chromosome Translocations 112
12. Complex Rearrangements 212 13. Parental Sex Chromosome Aneuploidy 221 14. Parental Autosomal Aneuploidy 233 15. The Fragile X Syndromes 242
7. Robertsonian Translocations 140
•
xiii
PART THREE: VARIANTS
PART SEVEN: PRENATAL DIAGNOSIS
16. Variant Chromosomes and Abnormalities of No Phenotypic Consequence 257
24. Parental Age Counseling and Screening for Fetal Trisomy 403
17. Copy Number Changes 269 PART FOUR: NORMAL PARENTS WITH A CHROMOSOMALLY ABNORMAL CHILD
18. Down Syndrome, Other Full Aneuploidies, and Polyploidy 277 19. Structural Rearrangements 295 20. Chromosomal Disorders of Sex Development 333 21. Chromosome Instability Syndromes 341
25. Prenatal Diagnostic Procedures 417 26. Preimplantation Genetic Diagnosis 426 27. Chromosome Abnormalities Detected at Prenatal Diagnosis 439 PART EIGHT: NOXIOUS AGENTS
28. Gonadal Cytogenetic Damage from Exposure to Extrinsic Agents 489 APPENDIXES
A. Ideograms of Human Chromosomes, and Haploid Autosomal Lengths 499 PART FIVE: DISORDERS ASSOCIATED WITH ABERRANT GENOMIC IMPRINTING
22. Uniparental Disomy and Disorders of Imprinting 351 PART SIX: REPRODUCTIVE FAILURE
23. Gametogenesis and Conception, Pregnancy Loss and Infertility 377
xiv
•
CONTENTS
B. Cytogenetic Abbreviations and Nomenclature 505 C. Determining 95 Percent Confidence Limits, and the Standard Error 510 References 511 Index 619
PART ONE BASIC CONCEPTS
This page intentionally left blank
1 elements of medical cytogenetics
CHROMOSOMES WERE first seen and named in the late nineteenth century. Chromosome is a combination of Greek words meaning colored (chrom) body (soma); the word was coined by the illustrious German anatomist Heinrich Wilhelm Gottfried von Waldeyer-Hartz. It was early appreciated that these brightly staining objects appearing in the cell nucleus must be the “stuff of heredity,” the very vessels of our genetic inheritance. Most observers had concluded, in the earlier part of the twentieth century, that the human chromosome count was 48. It was not until the 1950s, due to technical advances, and in particular the use of a hypotonic solution to swell the cells, giving an uncluttered view of the chromosomes, that Joe Hin Tjio and Albert Levan could recognize that 46 was the correct number. This discovery spurred research into conditions in which a chromosomal cause had hitherto been suspected; and in
1959 (“the wonderful year of human cytogenetics”) came the first demonstrations of a medical application of the new knowledge, with practically simultaneous discoveries of the chromosomal basis of Down syndrome, Klinefelter syndrome, and of Turner syndrome (Lejeune et al.,1 1959; Jacobs and Strong, 1959; Ford et al., 1959), and these were followed shortly thereafter by the recognition of the other major aneuploidy syndromes. Harper (2006) records the history, and the personalities behind the history, in his book First Years of Human Chromosomes; a book that should be read by every student of medical cytogenetics with an interest in how their discipline came to be. Harper points out that the practice of genetic counseling came into its own essentially upon the basis of these chromosomal discoveries: so to speak, geneticists now had “their organ.”
1 Among the “al.” was Marthe Gautier, who recounted, half a century following this report, her own less than fully acknowledged role in the endeavor; and Sir Peter Harper, in a commentary, and in his role as historian to the genetics community, takes an interpretative perspective upon this pioneering discovery (Gautier and Harper, 2009).
•
3
“Colored bodies” became an especially apt derivation with the development of various different staining techniques in the 1980s and 1990s, showing different parts of chromosomes in many different colors, whether true or computer-generated false colors. The images produced by this kaleidoscopic karyotyping could be rather beautiful. Black-andwhite photographs are less splendid but often suffice (Fig. 1–1). Albeit that molecular methodologies are now taking over from classical cytogenetics, and providing a different view of the genetic material, the word chromosome will surely last forever.
Chromosomal Morphology Chromosomes have a linear appearance: two arms that are continuous at the centromere. Reflecting the French influence in the establishment of the cytogenetic nomenclature, the shorter arm is designated p (for petit), and the longer is q (variously explained as being the next letter in the alphabet, a mistyping of g (for grand), or as the other letter in the formula
p + q = 1). In the early part of the cell cycle, each chromosome is present as a single structure, a chromatid, a single DNA molecule. During the cell cycle (Fig. 1–2) the chromosomes replicate, and two sister chromatids form. Now the chromosome exists as a double-chromatid entity. Each chromatid contains exactly the same genetic material. This replication is in preparation for cell division so that, after the chromosome has separated into its two component chromatids, each daughter cell receives the full amount of genetic material. It is during mitosis that the chromosomes contract and become readily distinguishable on light microscopy. (At other times in the cell cycle, chromosomes are attenuated and not visible as such.) Routine classical cytogenetic analysis is done on mitotic cells, usually obtained from blood. Blood lymphocytes have two convenient properties for the cytogeneticist: they are easily obtained, and they are easily stimulated to go into mitosis. The chromosomes of the small number of lymphocytes studied are taken as representative of the chromosomal
FIGURE 1–1 Banded chromosomes as they appear viewed through the microscope. 4
•
BA SIC CONCEPTS
Single chromatid chromosome
FIGURE 1–2
Replication
Separation at centromere
Daughter cells
Chromosome replication and separation during the mitotic cycle.
constitution of (essentially) every other cell of the body. Blood (specifically, nucleated white cells) is also the tissue from which DNA is extracted in routine microarray analysis. In the case of prenatal diagnosis, the cells from amniotic fluid or chorionic villi are the source material for both karyotyping and microarray analysis; these tissues are assumed (with certain caveats) to represent the fetal chromosomal constitution. The 46 chromosomes come in 23 matching pairs and constitute the genome. One of each pair came from the mother, and one from the father. For 22 of the chromosome pairs, each member (each homolog) has the same morphology in each sex: these are the autosomes. The sex chromosome (or gonosome) constitution differs: the female has a pair of X chromosomes, and the male has an X and a Y chromosome. The single set of homologs—one of each autosome plus one sex chromosome—is the haploid set. The haploid number (n) is 23. The haploid complement exists, as such, only in the gametocytes (ovum and sperm). All other cells in the body—the soma— have a double set: the diploid complement (2n) of 46. If there is a difference between a pair of homologs,
FIGURE 1–3
Segregation of chromosomes
in the sense of one being structurally rearranged, the person is described as a heterozygote. The chromosomes are classically distinguishable on the basis of their size, centromere position, and banding pattern. The centromere may be in the middle, off-center, or close to one end—metacentric, submetacentric, and acrocentric, respectively. The chromosomes are numbered 1 through 22, and X and Y, and are also assigned to groups A through G, according to their general size and the position of the centromere. The diagrammatic representation of the banding pattern is the ideogram (Appendix A). The numbering is based on size, largest to smallest (to split hairs, this order is not exact; for example, chromosomes 10 and 11 are shorter than chromosome 12, and chromosome 21 is smaller than 22). Certain parts of some chromosomes may show variation (heteromorphism) in the population. Increasing precision in banding permitted progressively more subtle definition of the chromosome (Fig. 1–3); microarrays take this to a further level. Chromosomes are conventionally displayed cut out from a photograph or captured from an electronic image, and arranged as a “paste-up,” with p arms
Increasing resolution of banding (chromosome 11). (Courtesy D. R. Romain.) Elements of Medical Cytogenetics •
5
upward, in their matching pairs. This paired-up presentation is called a karyotype, or karyogram (Fig. 1–4); the word karyotype is also used in the general sense of “chromosomal constitution.” Those coming from a DNA-based view may see the chromosome lying on its side, and microarray reports usually show a horizontal depiction of the chromosome arms, with the graph indicating duplications and deletions by a rise or a fall compared to baseline, respectively (although no one is proposing that short and long arms be renamed as left and right!). Karyotypes are described according to a shorthand notation, the International System of Human Cytogenetic Nomenclature (ISCN, 2009); an outline is given in Appendix B.
Chromosomal Structure and Function The two chemical components of chromatin are DNA and protein. Some of the stains used to produce chromosome banding patterns stain the DNA, while others stain the proteins. Proteins provide the scaffolding of the chromosome and are divided into histone and non-histone proteins. Histones are
1
6
2
7
13
14
19
20
strongly conserved DNA-associated proteins; the fact that they differ little between species such as ourselves and the sweet pea (for example) indicates how fundamentally important is their role in maintaining the integrity of the chromosome. Chromatin exists in differently condensed forms: the less condensed euchromatin and the more condensed heterochromatin. Euchromatin contains the coding DNA—the genes—while heterochromatin comprises noncoding DNA. Chromosomes are capped at the terminal extremities of their long and short arms by telomeres, specialized DNA sequences comprising many repeats of the sequence TTAGGG, that can be thought of as sealing the chromatin and preventing its fusion with the chromatin of other chromosomes. The centromere is a specialized region of DNA that, at mitosis, provides the site at which the spindle apparatus can be anchored and draw each separated chromatid to opposite poles of the dividing cell. Centromeric heterochromatin contains “satellite DNA,” so-called because these DNA species have different buoyant densities and produce distinct humps on a density gradient distribution. (These are not to be confused with the satellites
3
8
4
9
6
•
BA SIC CONCEPTS
10
11
12
16
17
18
15
FIGURE 1–4 Chromosomes arranged in formal karyotype.
5
21
22
X
Y
on acrocentric chromosomes.) A separate issue is the “packaging question”: how the centimeters of DNA are compacted into micron-length chromosomes. The presently preferred model is that the chromatin fibers are thrown into loops extending outward from a backbone, this backbone being formed as adhesive sites dotted along the fibers come together and construct a continuous linear stack. Miller and Therman (2001) treat this question in detail, and Annunziato (2008) provides a popular source.
CHROMOSOME ABNORMALITY Chromosomes are distributed to each daughter cell during cell division in a very precise process— precise, but prone to error. From our perspective, the two cell divisions of meiosis, during which the gametes are formed, are of central importance. Most of the discipline of medical cytogenetics focuses on the consequences of disordered meiosis having produced a chromosomally abnormal gamete, causing a chromosomal abnormality in the conceptus. A chromosome abnormality that is present from conception and involves the entire body is a constitutional abnormality. If an additional cell line with a different chromosomal complement arises before the basis of the body structure is formed (i.e., in embryonic or pre-embryonic life) and becomes an integral part of the organism, constitutional mosaicism results. In this book, we concern ourselves practically solely with constitutional abnormalities. Acquired chromosomal abnormality of course exists, and indeed it is a major initiating and sustaining cause in most cancers, a fact first proposed by Boveri in 1914, and voluminously attested by the work of Mitelman et al. (2011); but this is more the field of study of the molecular oncologist than the genetic counselor. An incorrect amount of genetic material carried by the conceptus disturbs and distorts its normal growth pattern (from zygote → blastocyst → embryo → fetus). In trisomy there is three of a particular chromosome, instead of the normal two. In monosomy only one member of the pair is present. Two of each is the only combination that works properly! It is scarcely surprising that a process as exquisitely complex as the development of the human form should be vulnerable to a confused outflow of genetic instruction from a nucleus with a redundant or incomplete database. Trisomy and monosomy for a whole chromosome were the first cytogenetic mechanisms leading
to an abnormal phenotype to be identified. More fully, we can list the following pathogenetic mechanisms that arise from chromosomal abnormalities: (1) A dosage effect, with a lack (deletion) or excess (duplication) of chromosomal material, whether for a whole chromosome or a part of a chromosome (Figure 1–5) (2) A direct damaging effect, with disruption of a gene at the breakpoint of a rearrangement (3) An effect due to the incongruent parental origin of a chromosome or chromosomal segment (genomic imprinting) (4) A position effect, whereby a gene in a new chromosomal environment functions inappropriately (5) Combinations of the aforementioned We discuss these mechanisms in more detail in following chapters.
Autosomal Imbalance STRUCTURAL IMBALANCE
As noted earlier, imbalance may involve the gain or loss of a whole chromosome—full aneuploidy—or of part of a chromosome—partial aneuploidy. The abnormality may occur in the nonmosaic or mosaic state. Loss (i.e., monosomy) of chromosomal material generally has a more devastating effect on growth of the conceptus than does an excess of material (i.e., trisomy). Certain imbalances lead to certain abnormal phenotypes. The spectrum is listed in outline in Table 1–1 and in more detail in Table 1–2; the spectrum with respect to mental retardation is depicted in Figure 1–6. Most full autosomal trisomies and virtually all full autosomal monosomies set development of the conceptus so awry that, sooner or later, abortion occurs—the embryo “selfdestructs” and is expelled from the uterus. This issue is further explored in Chapter 23. A few full trisomies are not necessarily lethal in utero, and many partial chromosomal aneuploidies are associated with survival through to the birth of an infant. Characteristically, “survivable imbalances” produce a phenotype of widespread dysmorphogenesis, and there may be malformation of internal organs and limbs. It is often in the facial appearance (facies) that the most specific physical abnormality is seen. The most complex organ of all, the brain, is the most vulnerable to a less than optimal genetic constitution; and some compromise of mental and intellectual Elements of Medical Cytogenetics •
7
Table 1–1. The Spectrum of Effects, in Broad Outline, Resulting from Constitutional Chromosomal Abnormality 1. 2. 3. 4. 5.
Devastation of blastogenesis, with transient implantation or nonimplantation of the conceptus Devastation of embryogenesis, with spontaneous abortion, usually in the first trimester Major disruption of normal intrauterine morphogenesis, with stillbirth or early neonatal death Major disruption of normal intrauterine morphogenesis, but with some extrauterine survival Moderate distortion of normal intrauterine development, with substantial extrauterine survival and severe mental retardation 6. Mild distortion of normal intrauterine development, with substantial extrauterine survival, and considerable intellectual compromise 7. Minimal physical phenotypic effect, varying degrees of intellectual compromise; possible compromise of fertility 8. No discernible physical phenotypic effect; cognitive function within the normal range, but less than expected from the family background
Table 1–2. The Impact of Constitutional Chromosomal Abnormality on Human Mortality and Morbidity, According to Classical Cytogenetics CONCEPTUSES OR INDIVIDUALS WITH
PROPORTION WITH CYTOGENETIC ABNORMALITY
“Occult abortion” (early embryonic death in unrecognized pregnancies) Miscarriage (recognized embryonic and fetal death (≥5 weeks gestation)
Unknown, perhaps a quarter to a half
Infant and childhood deaths Structural congenital malformations Congenital heart defects Multiple (three or more) birth defects and mental retardation Mental retardation (excluding fragile X)
About 30% total. Rate varies from 50% at 8–11 weeks to about 5% in stillbirths (≥28 weeks) 5%–7% 4%–8% 13% 5.5% IQ < 20 IQ 20–49 IQ 50–69
Other neurodevelopmental disability Criminality (defined by presence in security setting) Males in “ordinary” prisons Psychopaths, retarded criminals (male) Females in prison Male infertility (13% in those with azoöspermia) Defect in sexual differentiation (male) Ovotesticular disorder of sex development Defect in pubertal development (female) Primary ovarian deficiency Multiple miscarriage ? indicates a less certain estimate. Source: From Hook (1992).
? 3%–10% 12%–35% ? 3% ? 1%–3% 0.8% 3.0% 0.4% 2% <25% 25% 27% 65% 2%–5%
Dosage = 2 Two copies Normal
Dosage = 1 One copy, deletion Haplo-insufficient
Dosage = 3 Three copies, duplication Excess
FIGURE 1–5 Outline of normal chromosomal dosage (two copies), versus deletion (one copy), and duplication (three copies).
functioning, usually to the extent of an obvious deficit, is nearly invariable. While the physical phenotype in some cases of subtle deletion or duplication may be rather “bland,” compromise of neurological functioning is typical (Curry et al., 1997). Indeed, with the advent of microarray analysis, developmental delay or mental retardation2 in the absence of dysmorphism is becoming recognized as a chromosomal phenotype (Shevell et al., 2003; Shaffer et al., 2005; Macayran et al., 2006; Hochstenbach et al., 2009). The behavioral phenotype of autism is a frequent concomitant of imbalances at both the classical and microarray level (Rosenfeld et al., 2010a; Betancur, 2011). Thus, the central concern of most people seeking genetic counseling for a chromosomal condition is the fear of having a child who might have a physical, intellectual, or severe social handicap. FUNCTIONAL IMBALANCE
A correct amount of chromatin does not necessarily mean the phenotype will be normal. Inappropriate inactivation, or activation, of a segment of the genome can distort the genetic message. Some segments of the genome require only monosomic expression, and the homologous segment on the other chromosome is inactivated. If this control
fails, both segments can become activated, or both inactivated; and the over- or underexpression of the contained loci can cause phenotypic abnormality. The classic example of this is genomic imprinting according to parent of origin, and we discuss this concept in Chapter 22. A rather specialized example arises with the X-autosome translocation. A segment of X chromosome can fail to be inactivated; or conversely, X-inactivation can spread into an autosomal segment (Chapter 6).
Sex Chromosomal Abnormality Sex chromosome (gonosome) imbalance has a much less deleterious effect on the phenotype than does autosomal aneuploidy. The X chromosome is one of the larger and is gene-dense; the Y is small, comprising mostly heterochromatin, and carries very few genes. In both male and female, one, and only one, completely functioning X chromosome is needed. X chromosomes in excess of one are almost always inactivated, as the normal 46,XX female exemplifies. With X chromosome excess or deficiency, a partially successful buffering mechanism exists whereby the imbalance is counteracted in an attempt to achieve the same effect as having a single active X. In such states as, for example, XXX, XXY, XXXX, XXYY, and XXXXX, excess X chromosomes
2 Words can be powerful, and choice of language can help, or hinder, a counseling consultation: facts are to be conveyed clearly but also sensitively. The reader will have noticed our use of the expression “mentally retarded” in a number of places in this chapter. Some may have flinched; others may simply have accepted this as an accurate description. “Developmental delay” is a widely used term, and it can be perfectly appropriate in a pediatric setting, although less so in dealing with an adult. In the introduction to their paper on array analysis and karyotyping, Hochstenbach et al. (2009) refer to “idiopathic developmental delay (in infants <3 years of age) and mental retardation (in older children)”; and this distinction acknowledges that prediction of intellectual capacity is more precise in older children. And yet “mental retardation” has acquired a pejorative and somewhat harsh sense over the years, and some will prefer to use such expressions as “intellectual disability” or “cognitive compromise.” As we write elsewhere, counselors will need to know to whom they speak, and what language is best to use.
Elements of Medical Cytogenetics •
9
are inactivated. In the 45,X state, the single X remaining is not subject to inactivation. If an abnormal X chromosome (e.g., an isochromosome, or a deleted X) is present, then, as a rule, cells containing this abnormal chromosome as the active X are selected against, perhaps due to preferential growth of those cells in which it is the normal X that is the active one. In X imbalance, the reproductive tract and brain are the organs predominantly affected. The effect may be minimal. As for Y chromosome excess, such as XYY, there is a rather limited phenotypic consequence, but again the brain may be a vulnerable organ.
The Frequency and Impact of Cytogenetic Pathology According to the window of observation, chromosomal disorders make a greater or lesser contribution to human mortality and morbidity. Looking at prenatal existence, the earliest window has been provided by the in vitro fertilization (IVF) clinic, from the procedure of preimplantation genetic diagnosis (Chapter 26), at which single cells taken from 3-day-old embryos are subjected to genetic analysis; and an extraordinary fraction are chromosomally abnormal. After implantation (about day 5), and through the first trimester of pregnancy (to week 13),
chromosomal mortality is very high, and aneuploidy is the major single cause of spontaneous abortion (Chapter 23). Perinatal and early infant death has a significant chromosomal component, of which trisomies 18 and 21 (although the latter less so in more recent times) are major elements. As for morbidity, chromosomal defects are the basis of a substantial fraction of all intellectual deficit, and many of these retarded individuals will also have structural malformations that cause functional physical disability. Among a mentally retarded population, Down syndrome is the predominant contributor in the fraction who have a chromosome abnormality, while the increasing ability to pick up subtle deletions puts this category in second place (Fig. 1–6). Adolescence is a period in which many sex chromosome defects come to light, when pubertal change fails to occur; and in young adulthood, chromosomal causes of infertility are recognized. Few new cytogenetic defects come to attention later in adult life, but many retarded children survive well into adulthood and some into old age, and some require lifelong care from their families or from the state. This latter group imposes a considerable emotional and financial burden. While some parents and caregivers declare the emotional return they have from looking after these individuals, for others this
Down syndrome
76%
Other autosomal aneuploidy 1% Sex chromosome aneuploidy
4%
Rings and Markers 2% Deletions including 8% microdeletions Duplications
2%
Balanced rearrangements
3%
Unbalanced translocations Other
3% 1%
FIGURE 1–6 The relative proportions of different cytogenetic categories in 835 karyotypically abnormal individuals of a mentally retarded population in South Carolina studied in 1989–1994 (Phelan et al., 1996). If the exercise were to be repeated now, a lesser fraction due to Down syndrome might possibly be expected, due to prenatal diagnosis and pregnancy termination; and a new category would be needed for microduplications and microdeletions detected on microarray cytogenetics. (Courtesy M. C. Phelan; reproduced with the permission of the Greenwood Genetic Center.) 10
•
BA SIC CONCEPTS
responsibility is a source of continuing, unresolved, if attenuated, grief. Hook (1992) has summarized the categories of cytogenetic pathology and their impact, and we have reproduced his synopsis in Table 1–2. In Table 1–3 we set out the birth incidences of the various categories of chromosomal abnormality; these data are from a Danish study, one of a number that have examined this question in the later decades of the twentieth century, with largely similar findings in each. Overall, around 1 in 135 liveborn babies have a chromosomal abnormality, and about 40% of these are phenotypically abnormal due to the chromosome defect. If we were to look at 5-day blastocysts, the fraction with abnormality might be close to a half. If we studied a population of 70-year-olds, we could expect to see very few individuals with an unbalanced autosomal karyotype. A good approximation to a “normal” adult population—normal phenotypically and reproductively—is that reported in Ravel et al. (2006a) (Table 1–4), a study of 10,202 French sperm donors of proven fertility, bringing together data from a number of reproductive clinics comprising the “Centre d’Étude et de Conservation des Oeufs et du Sperme,” over the period 1973– 2002. Donors had an upper age limit of 50 years into the 1990s, and 45 thereafter. We suggest that the somewhat lesser fractions of some of the balanced karyotypes among these men may reflect a diminished fertility due to these rearrangements, with some translocation carrier men thus ineligible to have been recruited as donors. The finer the cytogenetic focus, the greater the incidence: with the highest resolution banding and the application of molecular methodologies, a number of previously unrecognized defects would be included. It will be a task for the cytogenetic epidemiologist of this century to derive new estimates of cytogenetic abnormalities in our population. THE RESEA RC H AP PLI CAT I ON OF CYTOGEN ETIC PATH OLOGY
The phenotypes that result from chromosome abnormalities can point the way to discovery of the causative genes. An early example of deletion mapping is the recognition that the gene for retinoblastoma was on chromosome 13, given the association of this cancer with the 13q– syndrome. Another cancer gene to be similarly mapped was APC (adenomatous polyposis coli), following the observation of polyposis in an individual with mental retardation
and del(5)(q22-q23). The triple dose of chromosome 21 in Down syndrome was a signpost on the way to finding the β-amyloid precursor protein (APP) gene as one of the Alzheimer disease loci. A translocation with one breakpoint at 7q11.23 was found to disrupt the elastin gene in a family segregating supravalvular aortic stenosis. Further investigation of this locus in Williams syndrome proved this to be the site of deletion in this condition (Nickerson et al., 1995). We have conducted a review of chromosomal conditions in which epilepsy is a feature, with the aim of providing leads to epilepsy genes (Singh et al., 2002a). The precision of microarray analysis, coupled with access to genome databases, now allows a much finer focus in the pursuit of causative genes. For example, Ou et al. (2008b) propose that one of the genes SIX1, SIX6, or OTX2 may be the basis of one form of branchiooto-renal syndrome, from their study of a child with a duplication of 14q22.3-q23.3. It is a general principle that many important scientific discoveries are made serendipitously; or, as Louis Pasteur put it, “chance favors the prepared mind” (le hasard ne favorise que les esprits préparés). Voullaire et al. (1993) identified a small supernumerary marker chromosome (sSMC) in a child with a nonspecific picture of physical abnormality and intellectual deficit, which had no C-band positive centromere (only a constriction). Conventional wisdom has it that a chromosome cannot be stably transmitted at cell division if it has no centromere. These workers studied this sSMC and discovered that it did have a simple, but nevertheless functional centromere. This observation led the way to the delineation of the “neocentromere” (p. 307). This elemental structure could be used as the basis for designing an artificial centromere, a necessary component of a human artificial chromosome (HAC). HACs may have a potential medical role as vectors for therapeutic genes. Historically, the chromosomal basis of some clinical syndromes has been identified following high-resolution analysis of cohorts of patients with similar phenotypes, or through the serendipitous identification of a visible chromosome abnormality. This “phenotype-first” approach led to the identification of many of the classical microdeletion syndromes. However, with the advent of microarray analysis, new syndromes are being identified based on their DNA aberration (Shaffer et al., 2007a,b). This “genotype-first” approach has led to the discovery of a number of new microdeletion syndromes, Elements of Medical Cytogenetics •
11
Table 1–3. Chromosomal Rearrangements and Imbalances, Recorded in 34,910 Live Newborns in Århus, Denmark, over a Total 13-Year Period, 1969–1974 and 1980–1988 NO. OF CASES
PER 1,000a BIRTH FREQUENCY PER GROUP
Sex Chromosomes Klinefelter Syndrome and Variants 47,XXY 47,XXY/46,XY 46,XX
20 7 2
1.12b 0.39 0.11
XYY 47,XYY 47,XYY/46,XY
18 2
1.01 0.11
XXX 47,XXX
17
1.00
1 3 1 1 2
0.06 0.18 0.06 0.06 0.12
Other 45,X/46,XY 46,XX/47,XX,del(Yq) 46,XX/46,XY Total
1 1 1 77
0.06 0.06 0.06 2.21
Autosomes Unbalanced Forms Trisomy 13 Trisomy 18 Trisomy 21 Trisomy 8 Supernumerary marker, ring Deletions, duplications
2 7 51 1 25 6
0.06 0.20 1.46 0.03 0.72 0.17
Turner Syndrome and Variants 45,X 45,X/46,XX and 45,X/47,XXX 45,X/46,X,r(X) 45,X/46,X,i(Xq)/47,X,i(Xq),i(Xq) Other Turner variant
1 in 616 4
1 in 894 4 1 in 1,002 5
1 in 2,130 5
1 in 453
1 in 379 Balanced Forms Robertsonian 13/14 translocation Other Robertsonian Reciprocal translocations Inversions (other than of chromosome 2) Combined sex plus autosomal totals Combined totals, excluding balanced autosomal forms
34 9 50 4
0.97 0.26 1.43 0.11
266 169
7.62 4.84
1 in 360 1 in 131 1 in 207
Notes: Not included in the 34,910 live newborns listing are four cases of sex chromosome induced abortion, involving the karyotypes 47,XXY, 47,XYY, 47,XXX, and 45,X/46,X,del(Xq), and 15 cases of autosomal induced abortions, involving the karyotypes +21, +13, +18, and three different derivative chromosomes. Had these pregnancies proceeded to term, the frequencies in the relevant group category would have been marginally increased. These figures might continue to be valid into the 2000s, except that the category of deletions and duplications will substantially increase, due to the more powerful detection now offered by microarray technology. a Per 1,000 male, per 1,000 female, or per 1,000 both, as appropriate. The gender-specific denominators in this study were 17,872 males and 17,038 females. b An increasing incidence of XXY in recent years has been suggested, and an Australian study, including data up to 2006, arrived at a figure of 1.91 per thousand (Herlihy and Halliday, 2008; Morris et al., 2008; Herlihy et al., 2010). Source: From Nielsen and Wohlert (1991).
Table 1–4. Chromosomal Rearrangements and Imbalances, at a 400–500 Band Level, Recorded in a France-Wide Study of 10,202 Fertile Men (Sperm Donors) over a 30-Year Period, 1973–2002
Robertsonian 13/14 translocation Reciprocal translocations Autosomal inversionsa 47,XYY 47,XYY/46,XY 47,XXY/46,XY b Supernumerary markerc Total
NO.
PER 1,000
7
0.67
5 5 5 1 2 4 29
0.5 0.5 0.5 0.1 0.2 0.4 2.84
a Excluding three cases of the common inv(2) and one of the common inv(10)(p11.2q21.2). b Excluding four cases of low level mosaicism, judged likely to have reflected cultural mitotic errors. c Including two cases of der(15) marker and one of iso(Yp) mosaicism. Source: From Ravel et al. (2006a).
representative examples of which are reviewed in Chapter 19. In the genotype-first approach, the phenotypes may vary considerably, making it difficult to describe a distinctive clinical syndrome. A similar variability is being uncovered in some of the classical syndromes, as the wider net that can be cast due to microarray brings in atypical patients for whom targeted fluorescence in situ hybridization (FISH) would not have seemed appropriate. With the use of whole genome scans, new syndromes are being identified, unappreciated clinical variation of classic syndromes is being uncovered, and the chromosomal etiology of some known syndromes is being discovered.
ETHICAL AND COUNSELING ISSUES Our focus in this book is on the biology of chromosomal defects and the reproductive risks they may entail. Certain bioethical issues, coming to be more formally defined in the late twentieth century, do, however, demand attention. Counselors must hold
fast to these requirements: (1) that they act beneficently toward their patients;3 and (2) that they strive to make their services accessible to those who may need them. N OND I R ECT I V E CO UN SEL I N G
In a Western ethos, the counselor is required to respect the autonomy of the client, and this largely translates into the principle that counseling be nondirective. Counseling may in fact never be truly nondirective, and we need to have an awareness of our own biases in order that our advice will be, as seen by those to whom we give it, valid. Rentmeester (2001) comments that, since it is “impossible for human language to convey facts purely, without any spoor of values,” and since “risk cannot be appreciated without consideration of values,” it is neither helpful nor possible to try to be value-neutral. There is a fine line between directive and detached counseling, a point nicely illustrated in Karp’s (1983) deft essay “The Terrible Question” (required reading for every counselor). Ingelfinger (1980) comments, admittedly in a somewhat different context: “A physician who merely spreads an array of vendibles in front of the patient and then says, ‘Go ahead and choose, it’s your life,’ is guilty of shirking his duty, if not of malpractice.” Rentmeester offers the refreshing advice that it is not necessarily unprofessional to answer a patient’s question: “What would you do?” It is the skill of the counselor that helps clients to reach the decision that is, for them, the right one, and for the clients to feel satisfied that they have done so. The subtleties and complexities of attempting to be nondirective in the setting of a prenatal diagnosis clinic are discussed by Anderson (1999), who analyzes responses of couples who did or who did not choose to have testing. She emphasizes the wide range of beliefs and values that people can have, as well as the likelihood for failed communication if these differences are not appreciated. In some other societies, the perceived good of the group may carry more weight than the professed wishes of the individual. The degree to which one society can seek to influence practice in another is a matter of some controversy, well illustrated by the response in the West to the “eugenic” Chinese Maternal and Infant Health Care Law of 1994
3 There seems no completely satisfactory word to use here, and we variably write of patients, clients, counselees, men and women, people, and “those whom we see.”
Elements of Medical Cytogenetics •
13
(Lancet editorial, 1995). The subtleties of the issue led to keenly pointed argument (correspondence in the American Journal of Human Genetics, 65, 1197– 1201, 1999). Knoppers (1998) comments upon the subtle boundary between the need to respect cultural, religious, and social diversity, and the imperative to adhere to tenets of generally accepted rights and ethics. More provocatively, she points to a “political and moral one-upmanship” which has colored the argument, and which may confuse deciding between what is “immoral state policy or just plain common sense.” TEST I NG C H ILD REN
To state the obvious, familial rearrangements are familial. It is very natural that parents would be concerned whether children they already have might be carriers, once an abnormality has been identified in one of them. Children, certainly, need to know their carrier status, sooner or later. It is very unfortunate (and possibly creates an exposure to legal redress) if a failure to transmit information leads to another affected child unknowingly being born elsewhere in the family. Burn et al. (1983) reported a family with a translocation having been the cause of cri du chat syndrome in two generations, the genetic information not having flowed through to the people who really needed to know it. We have had a similar experience: a family with a t(4;12) concerning which we had gone to the lengths of deriving and publishing a recurrence risk figure (Mortimer et al., 1980), and yet this information not traveling with a young man who had moved, as a child, from one country to another, and whose life has since been blighted by having had a daughter with partial 4p trisomy, and whose wife has had terminations due to unbalanced forms identified at prenatal diagnosis. On the other hand, genetic counselors are attuned to the principle of not taking away a child’s right to make, in the fullness of time, his or her own informed decision to learn about genetic risks he or she may face; thus, the principle is that the child’s future autonomy is to be respected. The American Society of Human Genetics and the American College of Medical Genetics (1995) have determined that “timely medical benefit to the child should be the primary justification for genetic testing in children and adolescents,” and it is true that a balanced chromosomal rearrangement will have no influence upon a person’s physical health, other than, in due
14
•
BA SIC CONCEPTS
course, his or her reproductive health (and the issue is thus to be seen in a different light than testing for adult-onset disease). Questions are raised that testing could damage a child’s self-esteem, distort the family’s perceptions of the child, and have adverse effects upon the child’s capacity to form future relationships (Clarke et al., 1994). Parents’ views are not without validity. Clayton (1995) comments that there is the possibility of conflict with parents, as physicians come increasingly to act as advocates for the child’s interests, but notes further that “children are generally ill-served if their parents feel they have not been listened to”; she also draws the conclusion that this is a medicoethical rather than a medico-legal issue. McConkieRosell et al. (1999) sought opinions from a group of 65 parents of fragile X children attending a national conference in Portland, Oregon, in 1996. They noted a “strong belief in a parental right to make the decision regarding carrier status in their children,” with about half considering that they should have the right to decide when their child should be tested and informed of the result. The Genetic Interest Group in the United Kingdom gently chided the profession in commenting that “the vast majority of people are better able to understand the implications than they are often given credit for” and has enunciated the following principle: “After suitable counseling, parents have the right to make an informed choice about whether or not to have their children tested for carrier status. Ideally, children should only be tested when of an age to be involved in the decision” (Dalby, 1995). It may be that earlier concerns overstated the potential for harm: at least with respect to the mendelian cancer-predisposing syndrome familial adenomatous polyposis, children having undergone predictive testing and receiving a positive gene test result experienced no increase in anxiety, depression, or loss of self-esteem (Michie et al., 2001). Indeed, Robertson and Savulescu (2001) see potential benefit to the child, and they support the view that, as a general rule, the parents’ views should prevail, and a request for predictive testing be respected. There is also the practical point that many parents will have had a prenatal karyotype from amniocentesis or chorionic villus sampling; and it may not seem entirely logical to decline to test a postnatal child. From the foregoing, we conclude that a conservative stance, but not an immovable one, is appropriate. Debating the issue with them, many parents
will see the wisdom of the declared position of the profession and be well satisfied (and possibly relieved) with the advice to leave testing until the child can decide. Equally, there will be occasions when acquiescence to a parental request may be reasonable. Either the parent’s mind is set at rest, or they know of the need to raise the issue with the child at a “suitable age,” which should be with the assistance of the genetic counseling clinic. The task for the counselor is to assist parents in deciding what age would be suitable for their child—“there is no universal ‘right’ age,” as McConkie-Rosell et al. (2002) comment with respect to fragile X carrier testing—and to convey the information in such a way that concern for the future is kept in perspective, and the child’s self-confidence is kept intact. And the pragmatic imperative: the wish to avoid family distress due to avoidable births of abnormal children in the next generation, as outlined earlier. Bache et al. (2007) found that 9% of carriers in Denmark, identified in childhood (or prenatally), had not been told as young adults; this observation led to a change in practice in that country, with a reminder letter being sent to the parents when their child reached the age of 18 years. FAM I LY STUD IES
More widely, the parents’ siblings and cousins could be carriers. Grandparental karyotypes may be useful in knowing which branch of a family to follow. The rights of individuals could, potentially, clash with the obligation that flows from belonging to a family: “no man is an island, entire unto himself ” and some may see altruism as a duty. Austad (1996) proposes that the family’s right to know about “sensitive genetic information” should take precedence over the individual’s right not to know. He considers it “alarming to use the principle of autonomy to renounce the co-responsibility for others, in this case, relatives”; and goes on to state that “we cannot exclude ourselves from the genetic fellowship of fate into which we are born.” If counselors take pains to provide clear information and to do so sensitively, such studies should usually proceed without unfortunate consequence. A suitable approach, in most families, will be to ask the person coming to the clinic to take the responsibility of bringing the matter to the attention of relatives, with appropriate support from the counselor. A letter couched in terms that it could be shown to other family
members, and providing contact points for further information, is often useful. Forrest et al. (2007) reviewed many international sources and identified these criteria seen as common obligations falling to the families, and to the counselors who see them: (1) individuals have a moral obligation to communicate genetic information to their family members; (2) genetic health professionals should encourage individuals to communicate this information to their family members; and (3) genetic health professionals should support individuals throughout the communication process. PRE D I CT I V E G EN E T EST I N G : DE L I B ER AT E A N D I N A DV ERT EN T
Counselors are very familiar with the concept of predictive genetic testing, that is to say, offering genetic testing to people who are presently well, but who are at risk for having inherited a particular genotype that may, at some stage in adult life, be the basis of the onset of disease. Its widest application is in the fields of cancer genetics and neurogenetics. With respect to translocations in the balanced state that may confer a predisposition to cancer, mention is made on p. 111, and over and above the reproductive implications of individuals being tested in such families, a cancer-associated risk will need to be assessed. As for inadvertent testing, we may mention a 30-year-old woman we have seen, presenting with premature ovarian failure and having a karyotype to check for an X chromosome mosaicism, but in whom trisomy 8 and a 14q;18q translocation were seen in 3/100 cells. She was otherwise in good health. This may well have been an “accidental” very early diagnosis of a lymphoma, and referral to a hematologist-oncologist—which was more than she had bargained for by having the test—was duly arranged. Concerning a neurogenetic focus, the delineation of the adult-onset neurodegenerative disorder associated with the fragile X premutation, and the premature ovarian failure in the female, add a layer of complexity to counseling issues with fragile X families. With the increasing application of microarray technology, the likelihood of discovering an incidental abnormality may now need more frequently to be taken into account, when a chromosome test is ordered. Schwarzbraun et al. (2009) report their experience in testing a severely mentally retarded and mildly dysmorphic 7-year-old girl, in whom
Elements of Medical Cytogenetics •
15
microarray revealed a de novo microdeletion (774 kb; contained 47 genes) at 17p13.1, and this deletion presumed to be the explanation for the clinical picture. One of the deleted 47 genes, however, happened to be TP53, and thus this deletion was considered to represent, effectively, a germline Li-Fraumeni4 mutation. This was quite unanticipated information for the parents to deal with, and the issue was further complicated by the child’s mental incapacity. Schluth-Bolard et al. (2010) consider this question, and they write “. . . the local Ethical Committee at the University Hospital of Lyon, France, suggested implementation of a plan to inform patients and their parents on the possibility of discovering pathology unrelated to mental retardation, and give them a month to carefully ponder on the possible consequences before signing the consent for study.” More pragmatically, they continue: “If this period of reflection would be difficult to apply in clinical practice, the possibility of incidental findings should be discussed during pre-test counseling and information should be given during post-test counseling by a trained clinician, aware of the potential psychological impact of such findings.” Netzer et al. (2009) tested a 5-year-old boy with mental retardation and identified a maternally inherited 24 kb intragenic deletion in the PARK2 gene, mutation in which is the basis of autosomal recessive juvenile Parkinson disease, and in which a risk to the heterozygote for typical late-onset Parkinson disease may also apply. These authors discuss the pros and cons of acceding to a parental request for having themselves tested in such a situation; there is no clear answer. We have dealt with the case of a child presenting with a clinical diagnosis of Beckwith-Wiedemann syndrome, in whom microarray revealed a maternally inherited 24 kb duplication at 11q12.2 that included the DAGLA gene. Elsewhere, we had proposed that duplication of this gene is a plausible candidate as the basis of spinocerebellar ataxia type 20 (Knight et al., 2008); thus, the question arose as to whether we might have inadvertently done a “predictive test” for this adult-onset neurological disorder, in the mother and child. The question was further nuanced by an uncertainty relating to the role of this gene, and thus whether
any further family testing would be a research versus a clinical exercise. “GU I LT ” I N A CA R R I ER
Sometimes a chromosomal diagnosis may be made in an older child or even an adult, where the parents will have held for years to the notion that obstetric misadventure, or a virus, or some other blameable event was the cause of the child’s condition. Some people find it upsetting to have to readjust, and to know that they may have been the source of the abnormality. They are likely to use words like guilt, blame, and fault. Helping these people to adjust to the new knowledge is a challenge for the counselor. They may eventually come to find the chromosomal explanation valuable and a source of some relief (as indeed some do at the outset). M E N TA L R ETA R DAT I O N A N D GE N ET I C A B O RT I O N
Intellectual deficiency is a condition for which many parents are unwilling to accept a significant recurrence risk—hardly remarkable, since intellectual function is such an obvious attribute of humanness. The great majority of those who chose to have prenatal diagnosis opt for pregnancy termination if a chromosomal condition implying major mental defect is identified. Some for whom abortion is not acceptable may nevertheless choose prenatal diagnosis for reassurance, or for the preparedness that certain knowledge can allow. Community views on mental handicap are changing and the late twentieth century saw something of an exodus from institutions and from special schools, as the mentally and psychologically disabled joined the “mainstream,” some more successfully than others. Many syndromes, in this Internet age, have their own support groups, and these are often a source of advocacy. Counselors need to handle the tension inherent in these views and the views of parents who want to avoid having a handicapped child; and the separate conflict that parents experience when a decision is taken to terminate an otherwise wanted pregnancy. As we discussed earlier, the doctrine of nondirective counseling is a central tenet of modern practice; and
4 Li-Fraumeni syndrome is a dominantly inherited cancer-predisposition syndrome, due to TP53 germline mutation, with severe implications. The cancers include, in early childhood, soft-tissue sarcoma, in later childhood, osteosarcoma, and in young adulthood, breast, brain, and hematological malignancy. It is controversial whether surveillance should be offered in childhood.
16
•
BA SIC CONCEPTS
it is a test of counselors’ professionalism that their own views not unduly influence the advice and counsel that they give. De Crespigny et al. (1998) document the experiences and comments of a number of couples in their book Prenatal Testing: Making Choices in Pregnancy, intended for the lay public. Walters (1995) and Tillisch (2001) offer personal perspectives. First, Walters: Defending the right of women who are carrying babies with Down’s syndrome to have abortions is not pleasant. Anyone who does so is likely to sound heartless, especially if they have no first-hand experience. It is even harder for me. I am the father of a Down’s syndrome baby. . . . It is the most painful thing I will ever say but my wife, Karen and I wish she had had a test. If she had, we would have terminated the pregnancy. I must be a callous swine, mustn’t I? . . . Her birth was a tragedy, but not so different to any tragedy that can strike out of the blue, such as a crippling accident. Just as we work to avoid other tragedies, I see nothing wrong in using Down’s tests to avoid the tragedy of human handicap. . . . I know that I would rather not have existed at all than to be, like her, sentenced to a life of confusion, frustration, pain and possibly loneliness when Karen and I are gone. If I feel guilt, it is that I was responsible for her birth. To me that guilt is far worse than anything I would have felt had I prevented it. Tillisch is the mother of a child with the del(1) (p36) syndrome (p. 311). Anomalies had been detected on ultrasonography during the pregnancy, but an amniocentesis returned a normal cytogenetic result. The child had a stormy neonatal course, and in due course the chromosomal defect was identified. Tillisch writes: I’m so thankful that the amniocentesis results were inaccurate. Since we didn’t learn of Kasey’s diagnosis until she was 9 months old, we were able to get to know, love, and admire Kasey as an individual, as our daughter. We didn’t allow doctors to define her for us. . . . From a mother’s perspective, Kasey’s future is bright. She receives treatment and will soon go to a public school. We will allow Kasey to show us her potential, rather than labeling her “severely mentally retarded” and casting her
off to be locked away from society. . . . My father once asked, if I could ever make Kasey “whole,” would I? Without any hesitation, I answered: absolutely not. Adding the missing genes would make Kasey a different person, a stranger. These differing, one could say polar views of parents find some parallels in the positions of those whom we could consider as the philosophers of our profession. Lejeune, in a provocative address to the American Society of Human Genetics in 1970, deplored the application of his original cytogenetic discovery to the prenatal diagnosis of Down syndrome. Epstein (2002) reflected, some three decades later, upon Lejeune’s influence, and while not stepping back from the standpoint that prenatal diagnosis is a proper and valid medical procedure, he does acknowledge (as must we) that a plurality of views exists, and that the genetics community must be sensitive to, and must respect, the range of views in the community. Brock (1995) discusses the philosophy of “wrongful handicap,” addressing the question of whether not producing a child who would suffer has harmed that potential child; and he enunciates a principle that “individuals are morally required not to let any possible child for whose welfare they are responsible experience serious suffering or limited opportunity if they can act so that, without imposing substantial burdens or costs on themselves or others, any alternative possible child for whose welfare they would be responsible will not experience serious suffering or limited opportunity.” This position could be seen as providing an ethically based framework for making a decision to terminate an abnormal pregnancy and to conceive again. PREG N A N CY A N D T H E M E NTA L LY R ETA R D ED
One issue to test the caliber of the bioethicist (not to mention the counselor) is that of the rights of the intellectually handicapped to have children (Elkins et al., 1986a). What of the person with Down syndrome, or some partial trisomy compatible with fertility, in whom a question of procreation arises? Zühlke et al. (1994) give an example in describing a man with Down syndrome who developed a relationship with a mentally retarded girl living in the same house. She requested removal of an intrauterine contraceptive device, became pregnant, and the Elements of Medical Cytogenetics •
17
normal baby is being brought up by the maternal grandmother. On the one hand, the right of the handicapped person to experience parenthood is debated; and the American Academy of Pediatrics (1990) expressed reservation about the sterilization of intellectually handicapped women on the basis of anticipated hardship to others. On the other hand, Gillon (1987) notes that normal people have the option of being sterilized, and the mentally handicapped should have the same right. The Law Lords in Great Britain concur that sterilization may be in the best interest of the handicapped person herself (Brahams, 1987). Many parents or guardians, not wishing to become “parental grandparents,” favor sterilization. Some regard hysterectomy as having the double benefit of ensuring sterility and facilitating personal hygiene; others consider only reversible contraception to be acceptable. The High Court of Australia decided in 1992 that the parents of a handicapped child cannot themselves lawfully allow sterilization, but that a court authorization is required, and noted that this requirement “. . . ensures a hearing from those experienced in different ways in the care of those with intellectual disability and from those with experience of the long term social and psychological effects of sterilization” (Monahan, 1992). Ten years later, it appeared that very few unlawful sterilizations of minors were being performed in the state of Victoria (Grover et al., 2002). When a retarded woman with a chromosomal defect is pregnant, or is pregnant by a retarded man, one or other of the couple having an unbalanced karyotype, and the pregnancy is recognized in time, the grounds for termination are substantial. The ethical issue arises over the difficulty (or impossibility) of securing the woman’s informed consent versus the expressed wishes of her guardians. Martínez et al. (1993) report from Alabama a mother with cri du chat syndrome, who was severely retarded and had no speech, pregnant by an unknown male, and “although pregnancy termination had been desired by the patient’s grandmother, social and legal limitations prevented access to this procedure.” Some less severely affected persons (if they are able to grasp the issue) may not regard it as undesirable to have a child like themselves; on the other hand, they may have the insight to recognize their own deficiency and not wish to pass it on. We may perhaps read this into the brief report of Bobrow et al. (1992) of a man with Down syndrome fathering a child, the mother having had first-trimester prenatal diagnosis 18
•
BA SIC CONCEPTS
(the baby was normal). There is the concept of imagining what a retarded person would want, were he or she intellectually competent to make a decision—a concept some would regard as paternalistic (and infringing personal autonomy) and that others see as valid and common sense. The sociology rather than the biology will exercise the counselor’s mettle in this uncommonly encountered situation. The other party is the child. Is having good parenting a right? What of a normal child born, say, to a man carrying a dup(10)(p13p14) chromosome and a mother with idiopathic mental defect? How can the interests of the child and of the parents be resolved? This is an actual case that we have seen (Voullaire et al., 2000a): it was quite poignant as this mildly retarded man, who had some insight into his own handicap, struggled to understand how best he might be a father to his 46,XX baby and expressed sadness at the abnormal behavior displayed by his older 46,XY,dup(10) child. The capable and willing grandmother stepped into the breach; but when the daughter is older, and assuming she is of normal intelligence, how will the realization of her parents’ abnormality affect her? Whether a normal child in this sort of setting has a legal claim for “dissatisfied life” is an intriguing and as yet (to our knowledge) untested notion (Pelias and Shaw, 1986). T HE STAT US O F EM B R YOS AT I N VI T R O F ERT I L I ZAT I O N
Lejeune has commented, indeed provided extensive testimony, on the ethical distinction between abortion and discarding an unwanted embryo. At a famous court case dealing with a dispute about IVF embryos in Blount County, Tennessee, in 1989, he insisted on the point that human life commences at conception, and therefore that disposing of a zygote is, in essence, no different from the induced abortion of an established pregnancy. This argument is not necessarily seen as convincing to those pragmatic couples who choose to have preimplantation diagnosis in order to avoid the predicament of having to decide upon a course of action following prenatal diagnosis of a chromosomal abnormality at chorionic villus sampling or amniocentesis. One Catholic thinker is of the opinion that “human personhood” of the embryo does not inhere until the stage at which embryonic cells have differentiated and the primitive streak has appeared (at about the end of the second week post-conception) (Ford, 1988). Prior to that time, when the “pro-embryo,” as
he prefers to call it, is only a personne en devenir, “we should resist the conceptual and linguistic temptation to attribute an unwarranted ontological unity to an actual multiplicity of developing human blastomeres.” More liberally, Isaacs (2002) discusses the concept of a continuum, in which the “moral status” of the fetus increases in value through pregnancy (and indeed after birth); and some couples seem intuitively to follow this line. These issues underlie arguments about the validity or not of the term “pre-embryo” ( Jones and Veeck, 2002; Tacheva and Vladimirov, 2002, et seq.) ACCESS TO PRENATA L D I AG N OSTIC SERV ICES
It would not, at present, be economically feasible or sensible to make definitive prenatal diagnosis (chorionic villus sampling or amniocentesis) available to every pregnant woman. Even among those for whom testing is, in principle, freely available, a proportion will not present, either because they are opposed to abortion, or because they have not been informed about, or have not understood, the issues involved (Halliday et al., 2001). Those who can afford it and who do not meet criteria (essentially maternal age or other particular indicators of risk) for acceptance in the public system may have the privilege of access to private testing. In the United Kingdom, the question has been raised that a wider availability, although at the expense of a lesser detection rate, would follow the implementation of a rapid diagnostic test targeting the major aneuploidies (qualitative fluorescent polymerase chain reaction [QF-PCR], FISH) (Mackie Ogilvie et al., 2009). Mass screening methodologies (Chapter 24) are to some extent bypassing the inequity inherent in the public/ private dichotomy. If noninvasive early prenatal diagnosis (Chapter 25), using such methodologies as the analysis of fetal DNA or RNA from a maternal blood sample, becomes available, then potentially all pregnancies could be subject to chromosomal analysis; but this ready availability would, of itself, raise a question about the need for satisfactory counseling prior to undergoing such an “easy” procedure as a venepuncture (Schmitz et al., 2009; de Jong et al., 2010).
Legal barriers may arise in some jurisdictions. In the United States, as Miller et al. (2000) comment, “there is perhaps no more divisive subject than abortion.” A possible ban on second-trimester (14–27 weeks) abortions would considerably affect couples having prenatal cytogenetic diagnosis, since many chromosomal abnormalities are discovered in the second trimester, and particularly in the period 14–18 weeks. Miller et al. calculated that a secondtrimester ban would have a net annual cost of $74 million in the state of Michigan, and $2 billion in the United States, based on the estimated lifetime costs of individuals with various congenital defects (including other than chromosomal). If prenatal testing is not made available, or if an abnormal result is reported but has not been passed on to the parents, the option of pregnancy termination is denied. Here, the legal concept of the “right not to be born” may be invoked (Weber, 2001). The issue is controversial.5 French courts made landmark decisions in 2000 and 2001 in which substantial financial compensation was granted to parents of children with Down syndrome. Whatever the legalities, the lesson for the counselor is that testing should be offered to those for whom it may be appropriate, and that they should be diligent and careful in ensuring that prenatal testing results are safely conveyed to the right person. PA RT I CI PAT I O N I N R ESEA R CH
There is much yet to learn about clinical cytogenetics, and much of this cannot be done without patient participation (a rather obvious statement, and one that applies to medicine generally). It is, of course, well enshrined that patients who are potential recruitees should be fully informed upon the implications for themselves of a study in which they might be invited to participate, and that they have the opportunity to decline, without compromise of their own health care. Having made that point, one can see a reciprocity in providing a health care service: the patient who benefits (possibly at the expense of the state) could be seen as having a moral duty at least to consider an invitation to be involved in a bona fide research study. And having made that point, the reality is that, rather often, patients are
5 A claim for “wrongful life” concerning cri-du-chat syndrome was brought on behalf of the child in a legal case in Australia, whose birth followed a failed vasectomy (Watson, 2002). The claim failed, the judge finding it impossible to compare, and to place values upon, impaired existence versus nonexistence.
Elements of Medical Cytogenetics •
19
very willing to come forward, and they gain some satisfaction in feeling that they may be making a contribution toward the greater good: the altruism gene shines brightly in many people. It was thus disappointing to read in Giardino et al. (2009) (and see p. 467), that a large study on de novo rearrangements detected at prenatal diagnosis could not be properly completed, in which data on a little over a quarter-million pregnancies were accumulated, from several Italian cities, and a good number (246) of de novo rearrangements identified. Here was an opportunity to build on the remarkable work of Warburton (1991). But, as these authors write, “Unfortunately, our limited information regarding the frequency and type of clinical features associated with the prenatal detection of apparently balanced rearrangements did not allow us to improve prenatal genetic counseling by updating the risk provided so far by Warburton.” One perfectly valid reason may have been the logistics: “ . . . the diagnostic laboratories, the services providing genetic counseling and follow-up and the hospitals where the births take place are not integrated, but often topographically [geographically] distant.” Organizing multicenter research, and undertaking
20
•
BA SIC CONCEPTS
field work to gather data, is certainly challenging. However, it appeared that privacy concerns trumped any other issue: “Furthermore, request of further information in the absence of a specific consensus is forbidden by the actual [present] privacy law.” And it did not escape notice that, in the same issue of Prenatal Diagnosis in which this paper appeared, another paper (Ramsay et al., 2009) examined the attitudes toward research participation of parents whose child had had an abnormality shown at prenatal ultrasound. To quote these authors: “ . . . the balance falls between the possibility of causing upset to parents, particularly those with handicapped or ill children, and the possibility of gaining new knowledge that may prove important to parents deciding whether or not to continue their pregnancy after diagnosis of a fetal abnormality.” Their study in fact demonstrated “that the great majority of respondents indicated they would be happy to be contacted to provide information on their children’s health and development . . . Research ethics committees can be reassured that the risk of causing inappropriate and unnecessary parental distress by inviting them to take part in such studies is low.”
2 chromosome analysis
ON CLASSICAL METHODOLOGY, chromosomes are analyzed in the cytogenetics laboratory under the light microscope at a magnification of about 1000×. The chromosomes must be stained to be visible, and a great many staining techniques have been used to demonstrate different features of the chromosome. We list some of these, in particular those with a more immediate practical application to the clinical issues we discuss in this book, or which are of historical value, when referring to the older literature.
CLASSICAL CYTOGENETIC ANALYSIS 1. Plain staining (“solid staining”). Many histologic dyes, including Giemsa, orcein, and Leishman, stain chromosomes uniformly. Until the early 1970s these were the only stains available. 2. Giemsa or G-banding. This procedure requires a trypsin (protein digestion) step and is the main staining method in use in routine classical
cytogenetics. It allows for precise identification of every chromosome and for the detection and delineation of structural abnormalities. At the 400– 550 band level, rearrangements down to about 5 megabases in length can be discerned, at least in regions where the banding pattern is distinctive. Its precision is increased by manipulations designed to arrest the chromosome in its more elongated state at early metaphase or prometaphase—high-resolution banding. Alternative methods to demonstrate essentially the same morphology are quinacrine or Q-banding and reverse or R-banding. In R-banded chromosomes the pale staining regions seen in G-banding stain darkly, and vice versa. 3. Constitutive or C-banding. This technique stains constitutive heterochromatin—mainly the centromeric heterochromatin, some of the material on the short arms of the acrocentric chromosomes, and the distal part of the long arm of the Y chromosome. Constitutive heterochromatin, by definition, has no direct phenotypic effect and, in general, is devoid of active genes. •
21
4. Replication banding. This technique is used primarily to identify inactive X chromatin. A nucleotide analog (BrdU) is added either as a pulse at the beginning, or toward the end of the cell cycle, to allow the cytogenetic distinction of chromatin that replicates early from that which replicates late. It produces a banding pattern similar to that of R-banding. 5. NOR (silver) staining. This stain, of largely historic interest now, identifies nucleolar organizing regions (NOR), which contain multiple copies of genes coding for rRNA, and are sited on the satellite stalks of the acrocentric chromosomes; these can also be demonstrated on FISH. 6. Distamycin A/DAPI staining. This fluorescent stain identifies the heterochromatin of chromosomes 1, 9, 15, 16, and Y. A particular use is to distinguish the inverted duplication 15 chromosome (p. 306) from other small marker chromosomes. 7. Fluorescence in situ hybridization (FISH) and variations thereupon. The major cytogenetic advance of the 1990s was the ability to identify specific chromosomes and parts of chromosomes by in situ hybridization with labeled probes. It is widely used to detect submicroscopic deletions and to characterize more obvious chromosome anomalies. The hybridization method may be direct or indirect. Direct attachment of a detectable molecule (e.g., a fluorophore) to the probe DNA enables its microscopic visualization immediately after its hybridization to the target DNA in the chromosome. The more sensitive indirect procedure requires special modification of the probe with a hapten detectable by affinity cytochemistry. The most popular systems are the biotin-avidin and digoxigenin systems. By using combinations of biotin-, digoxigenin-, and fluorophore-labeled probes, multiple simultaneous hybridizations can be done to locate different chromosomal regions in one preparation (multicolor FISH).
rearrangements not detectable on routine banded analysis. The frequency of subtelomeric abnormalities, identified by FISH in the cytogenetics laboratory, is estimated to be about 2.5% of individuals tested (Ravnan et al., 2006). Because the subtelomere FISH assay is labor intensive and limited to only the ends of the chromosomes, this analysis has been largely replaced by microarray-based analysis. 9. Comparative genomic hybridization (CGH). In CGH, differentially labeled, fluorophore-tagged DNA from the patient and a normal control (reference sample) is applied to a metaphase slide prepared from a “standard” normal person. Relative excesses and deficiencies of patient DNA bind competitively with respect to the control onto the reference chromosomes and yield different color intensities on exciting the fluorophores. This procedure has even been applied successfully to archival pathology material. It may have application in preimplantation genetic diagnosis, where it offers the advantage of testing the whole karyotype. “Highresolution” CGH refers not to a more stretched chromosome preparation, but to a further level of sophistication of the computer software that is used to analyze the images, by adjusting for the idiosyncratic patterns that each homolog may have. Small imbalances may be identifiable by this approach, ~10 Mb or greater, and the nature of uncertain rearrangements clarified (Knight & Flint, 2000; Kirchhoff et al., 2001; Ness et al., 2002). The ability to detect interstitial abnormalities offers an advantage over subtelomeric FISH (Kirchhoff et al., 2004).
A more focused use of FISH is in the assessment of imbalances revealed by microarray analysis (see later discussion), with the probe from the genomic region targeted to the specific region identified by the array.
MICROARRAY ANALYSIS
8. Submicroscopic telomeric analysis. The subtelomeric regions are, in general, gene rich, and very small rearrangements in these regions can have a profound effect. Probes have been developed for targeted subtelomeric FISH that may identify subtle 22
•
BA SIC CONCEPTS
Chromosomes examined by various techniques are illustrated in Figures 2–1. Full detail is to be found in Keagle and Gersen (1999), Blancato (1999), Mark (2000), and Miller and Therman (2001), while Trask (2002) provides a historical span of the cytogeneticist’s skill.
Having held sway for the better part of a halfcentury, classical cytogenetics is now yielding primacy of place to the power of molecular methodology, in the form of microarray analysis. Indeed in 2010, a consensus statement over the names of 32 geneticists from several countries was entitled ‘Chromosomal microarray is a first-tier clinical diagnostic test for individuals with developmental disabilities or congenital anomalies’ (Miller et al., 2010);
(a)
(b)
(c)
(d)
(e)
(f) 1
6
15
16
YX
FIGURE 2–1 Chromosome pairs 1, 6, 15, 16, and Y and X stained by various techniques: plain stain (a), G-banding (b), replication banding (c), C-banding (d), Ag-NOR stain (e), and Q-banding (f).
and from the Professional Practice and Guidelines Committee of the American College of Medical Genetics, ‘cytogenetic microarray analysis is recommended as a first-line test in an individual with multiple anomalies not specific to a well-delineated genetic syndrome, apparently nonsyndromic developmental
delay/intellectual disability, and autism spectrum disorders’ (Manning and Hudgins, 2010). Not that classical cytogenetics is likely to fade from view: there are two crucial reasons for its continuing use in the laboratory. First, not all array results can give a definitive construction, and FISH is often necessary Chromosome Analysis •
23
to elucidate the cytogenetics. Second, the array cannot detect balanced rearrangements, and recognition of the carrier state will continue to need an old-fashioned chromosome test. And third, a rather subjective “reason” is that, by continuing to work with chromosomes, the molecular cytogeneticist will not lose the intuitive understating of what chromosomes are really like, and not see them merely as theoretical constructs from which the DNA has been extracted. The reporting of microarray results is a sophisticated exercise, and counselors need to be sophisticated readers of these reports; many laboratories now use depictions from one of the genome browsers to illustrate the precise extent of the imbalance, and noting the genes contained within this segment. There are basically two types of microarrays available at this time: those that use a CGH approach, much like that described earlier for chromosomal CGH, and those that use single nucleotide polymorphisms (SNP) to assess the number of alleles in a sample. Although microarrays can differ in their genomic composition and substrates used for the analysis, most microarrays are comprised of thousands of spots of reference DNA sequences, applied in a precisely grided manner upon a slide (or “chip”) in which the locations can be known by computer analysis.
Comparative Genomic Hybridization The fundamental principle is essentially the same as in chromosomal CGH, noted earlier: using the array, rather than the metaphase spread, as substrate. Patient and control DNA are labeled in two different fluors, usually one that appears red and one that appears green. These labeled DNAs are applied to the microarray, and hybridization takes place. Typically, if the number of copies between the control and the patient are the same, the spot looks yellow (produced from an overlapping of equal amounts of red and green). The fluorescent intensities of each dye are measured. If the patient has an excess at a locus (due to duplication or aneuploidy), the hybridization will more reflect the dye of the patient’s DNA. If the patient has a deficiency at a locus (loss due to deletion or unbalanced translocation), the hybridization will more reflect the dye of the control DNA. These fluorescent intensities are presented as a ratio of each of the dyes and plotted as shown in Figure 2–2. Microarrays for CGH are typically constructed from bacterial artificial 24
•
BA SIC CONCEPTS
chromosomes (BACs) or oligonucleotides. Each spot represents a unique BAC or oligo. For example, an array with 3000 BAC spots could detect unbalanced rearrangements at a 1 Mb resolution across the entire genome (Snijders et al., 2001). The power of array-CGH over classical cytogenetics is illustrated in a study from Finland, in which ~20% of 150 patients with mental retardation, and whose G-banded karyotypes had previously been assessed as normal, showed a presumed pathogenic imbalance on microarray (Siggberg et al., 2010).
Single Nucleotide Polymorphism Like the microarrays described earlier for CGH applications, single nucleotide polymorphism (SNP) arrays can be used to detect the number of alleles in a specimen. Heterozygosity, with two distinct alleles, can be distinguished from homozygosity, and from three alleles. Apparent homozygosity may indicate a loss of DNA, such as a deletion, while three alleles may indicate a gain of DNA copy number, such as a duplication or trisomy. SNP-based microarrays have the added advantage of detecting uniparental disomy when the child’s results are compared to the parental genotypes. Isodisomy may be revealed, in the absence of parental samples, when the entire chromosome shows homozygosity and chromosomal monosomy is an incompatible interpretation. Balanced rearranged chromosomes, it is to be noted, cannot be detected using any of the current microarray-based technologies. The only exception, though it is not amenable to diagnostics currently, is the technique of array painting. This technology combines the use of flow sorted chromosomes to separate the two derivatives of a balanced translocation, amplifies the DNA, and applies each amplified derivative to a microarray to determine the breakpoint locations and size of the segments involved (Gribble et al., 2004). POLYM ER A SE CH A I N REACTION–BASED APPLICATIONS
A number of technologies are available to assess DNA copy numbers. These are targeted approaches to answer a specific question: How many copies of the target are present in the patient? These techniques include quantitative fluorescent polymerase chain reaction (QF-PCR) and multiplex ligationdependent probe amplification (MLPA). QF-PCR and MLPA use specific primers to amplify segments
FIGURE 2–2 Laboratory process for microarray analysis by comparative genomic hybridization. See also separate color insert.
of DNA to determine copy number and identify deletions or aneuploidy. Since telomeric imbalances may be relatively frequent, and often undetectable even by highresolution cytogenetics, approaches have also been devised to assess these regions by DNA-based methodologies instead of FISH or microarrays. They have the particular advantages that all subtelomeric regions are assessed in a single test, and that several samples can be run together as a batch. One approach is that of multiplex amplifiable probe hybridization (MAPH), in which genomic DNA is analyzed with a set of small (140–600 bp) probes for subtelomeric sequences from every chromosome, and the degree of amplification quantified (Hollox et al., 2002). Greater or lesser amplification indicates a duplication or deletion on that particular chromosome arm. “ NE X T- G EN ER ATIO N SEQU E NCI NG”
DNA methodologies based on massively parallel genomic (“next-generation”) sequencing have
enabled remarkable advances in mutation analysis, with the entire expressed genetic complement, the “exome”, tractable to interrogation. In the cytogenetic field, this approach is showing promise as a first trimester screening test for trisomy 21 that could potentially avoid invasive prenatal diagnosis, by measuring the amount of chromosome 21 DNA of fetal origin in the maternal plasma (see p. 423). Chromosomes, as viewed in this new century, are beginning to resemble the graph in Figure 2–3, and the interpretation will be based upon this sort of raw data. Although cytogenetics will continue to evolve, whatever techniques come to be used, the fundamental purpose of the cytogenetic report will of course remain the same. Descriptions about the technologies used will be important addenda to reports, because they may inform the clinician about the interpretation of the chromosome analysis, and the need for further possible analysis. Reports may also include a listing of presumed significant genes in the region, a comment upon imprinting, and the likelihood of benign versus causative genomic Chromosome Analysis •
25
FIGURE 2–3 Plot of chromosome 22 in a patient with a 22q11 deletion performed using an oligonucleotidebased array with comparative genomic hybridization. The deletion is indicated by the shaded area, which reflects a deviation from the log2 ratio of 1 (equal to zero). Distal to the classic diGeorge deletion is a common copy number variant (CNV).
changes. Regardless of the technology used, visualizing the karyotype by banding or FISH is, we propose, an essential part of the interpretation, and critical to enabling well-founded genetic counseling for the family (Bui et al., 2011).
GENETIC COUNSELING CONSIDERATIONS In the majority of cases, the abnormalities found by molecular technologies have clear clinical relevance for the patient. However, higher resolution strategies will uncover DNA changes of unclear clinical significance. Such findings may require testing of additional family members, parents, grandparents, and sometimes siblings, to understand the relationship, if any, between the DNA alteration and the clinical phenotypes or medical problems of the
26
•
BA SIC CONCEPTS
patient. The possibility of findings of unclear clinical significance should be discussed when ordering the test, especially in the prenatal setting. Because these molecular-based tests have the ability to interrogate the entire genome, the pretest genetic counseling should include information about uncovering unwanted information, such as loci that can predispose to cancer loci, or to adult-onset disorders. The use of SNP arrays may uncover substantial stretches of homozygosity due to consanguineous or even incestuous relationships (Schaaf et al., 2011). These counseling caveats notwithstanding, the higher resolution potential of these new technologies will make a significant impact on our ability to make diagnoses, will increase the detection rate of chromosome abnormalities, and substantially improve our ability to provide the answers that families seek.
3 the origins and consequences of chromosome pathology
“WHAT WENT WRONG? And will it happen again?” These are the common questions from “chromosomal families” that bring people to the genetic clinic. We can recast these questions: “Did I, or one of us, produce an abnormal gamete? If so, why? What gamete might be produced next time? Or, if the chromosomes were normal at conception, what went wrong thereafter?” To deal intelligently with these questions, the counselor needs a broad knowledge of how gametes form, how chromosomes behave, and how the early conceptus grows. We consider the distinction between abnormality due to structural defect (full or segmental aneuploidy), the majority, and the small fraction due to functional defect (aberrant imprinting status). Most of the chromosome abnormalities in individuals that counselors see in the clinic will have arisen from errors during formation of the germ cells, and we focus particularly upon meiosis, the specialized cell division of gametogenesis. Chromosome defects can arise postzygotically, and abnormalities of mitotic cell division in the cleavage-stage embryo
and in the embryo proper can produce chromosome mosaicism; we review the possible consequences of this. We refer in passing to the concept of dynamic mutation, but we leave its fuller discussion for the fragile X chapter (Chapter 15). First, we look at etiology. We discuss three chromosomal settings within which genetic abnormality may arise, namely meiosis, mitosis, and genomic imprinting. Within each, we consider what types of abnormality may happen. In meiosis and mitosis, irregular segregation can produce aneuploidy for a whole chromosome, while asymmetric segregation of a structural rearrangement produces an incorrect amount of part of a chromosome (partial, or segmental aneuploidy). In genomic imprinting, the defect is qualitative, with abnormal expression of what can be a normal amount of chromosome. Sometimes there is overlap: for example, a meiotic error can subsequently lead to an abnormality of imprinting. Sometimes we cannot be sure which is the correct category: a supposed meiotic error, for example, could actually have arisen in a premeiotic •
27
mitosis. Nevertheless, this format is not too arbitrary, and it provides a useful framework within which the generality of chromosomal abnormality can be appreciated. Second, we consider pathogenesis: the process by which the underlying genetic defect then leads to phenotypic abnormality. Third, and with particular reference to the question of recurrence risk advice, we make some general comments about which categories of abnormality are likely to recur, or for which sporadic occurrence is the rule. Spermatogenesis
Oögenesis
Gonadal stem cell
Replication
Primary gametocyte MEIOSIS I Secondary gametocyte
PB1
MEIOSIS II
PB2
PB1
Spermatids Mature ovum and polar bodies
Spermatozoa
FIGURE 3–1 Outline of chromosomal behavior and distribution during gametogenesis. Each primary spermatocyte divides symmetrically at the sequential meioses into four spermatids. Division of the öocyte is asymmetric as it buds off the first polar body (PB1) at meiosis I, and the second polar body (PB2) at meiosis II. (The first polar body may or may not divide at meiosis II; as shown here, it has not).
MEIOSIS Meiosis in Chromosomally Normal Persons The purpose of meiosis is to achieve the reduction from the diploid state of the primary gametocyte (2n = 46) to the haploid complement of the normal gamete (n = 23), and to ensure genetic variation in the gametes. The latter requirement is met by enabling the independent assortment of homologs (the physical basis of Mendel’s second law),1 and by providing a setting for recombination between homologs. While we do not dwell on recombination per se, this is, to the classical geneticist, a raison d’être of the chromosome: “from the long perspective of evolution, a chromosome is a bird of passage, a temporary association of particular alleles” (Lewin, 1994). The mature gamete is produced after the two meiotic cell divisions: meiosis I and meiosis II (Fig. 3–1). In meiosis I, the primary gametocyte (oöcyte or spermatocyte, also referred to as primordial germ cells) gives rise to two secondary gametocytes, each with 23 chromosomes. These chromosomes have not divided at the centromere, and they remain in the double-chromatid state. In meiosis II, the chromosomes of the secondary gametocyte separate into their component chromatids. In the male, the daughter cells produced are the four spermatids, which mature into spermatozoa. In the female, the daughter cells are the mature ovum and its polar bodies. (In fact, it is not until sperm penetration that meiosis II in the ovum is completed.) Each gamete contains a haploid set of chromosomes. The diploid complement is restored at conception with the union of two haploid gametes. The moment of conception, as the embryologist sees it, is not at sperm penetration, but only when the two pronuclei have fused to form a single nucleus (“syngamy”). Note that spermatogenesis divides the cytoplasm evenly, so that after meiosis II there are four gametes of equal size. The sperm head that penetrates the ovum comprises almost entirely nuclear material; the tail is cast off. In oögenesis, cytoplasmic division is uneven, producing a secondary oöcyte and first
1 The law of independent assortment: During gamete formation the segregation of the alleles of one allelic pair is independent of the segregation of the alleles of another allelic pair. The exception: If two loci are close together—“linked”—on the same chromosome.
28
•
BA SIC CONCEPTS
polar body after meiosis I, and the mature ovum and second polar body at meiosis II. The chromosomes of the first polar body may or may not undergo a second meiotic division (which would in any event be a pointless achievement). The ovum and its polar bodies each have a haploid chromosome set, but the ovum retains almost all of the cytoplasm.2 Another major sex difference concerns the timing of gamete maturation. In the female, meiosis is partway through, in the late prophase of meiosis I, by the eighth month of intrauterine life (the actual process of recombination takes place during weeks 16–19 of fetal life). At birth, on average there are somewhat over half a million oöcytes (Bukovsky et al., 2004). Most of this pool gradually disappears, but those eggs destined to mature stay in a “frame-freeze” until they enter ovulation, some one to five decades thereafter, and meiosis recommences. Testicular stem cells, on the other hand, do not begin to enter meiosis until the onset of puberty. Thereafter, millions of mature sperm are continuously produced. We now examine more closely the details of meiosis. During the final mitotic division in the primary gametocyte, the homologous pairs of chromosomes have (as with any mitosis) replicated their DNA to change from the single-chromatid to the double-chromatid stage. They now enter into the meiotic cell cycle (Fig. 3–2a). As meiosis I proceeds to prophase, chromosomes conduct an “homology search” and come together and pair, with matching loci alongside each other (Fig. 3–2b). This process—synapsis—continues with a more intimate pairing of the homologs, starting at the tips of the chromosomes and proceeding centrally (Barlow and Hultén, 1996), and the synaptonemal complex is formed. The paired chromosomes themselves are called bivalents.3 Synapsis sets the stage for an exchange of matching chromosome segments; this is the process of recombination, or crossing-over (Fig. 3–2c). Next, desynapsis occurs (the diplotene stage), with dissociation of the synaptonemal complex and the formation of chiasmata. Now, the
two homologous chromosomes disjoin and go to opposite poles of the cell. This is the anaphase stage; the orderly movement of chromosomes during this sequence is facilitated if synapsis, recombination, and chiasmata formation have proceeded normally. Finally, the cell divides into the two daughter cells (Fig. 3–2d). How the chromosomes are distributed—which chromosome goes to which pole—is called segregation. Normally, each daughter cell gets one of each of the pair of chromosomes, and this is referred to as 1-to-1 (1:1) segregation. Uniquely in the meiosis I cell division, daughter cells are produced with double-chromatid chromosomes. These cells then enter meiosis II (with the possible exception of the first polar body, as noted earlier). In this cycle, the chromosomes do not replicate, because they are already in the double-chromatid state. The chromosomes separate at the centromere, and the resulting single-chromatid chromosomes disjoin, one going to each pole, resembling a mitotic division (Fig. 3–2e). The course of meiosis is discussed in much fuller detail in Miller and Therman (2001). Chromosomal pathology arises when these processes of disjunction and segregation go wrong— nondisjunction and malsegregation.
Meiosis in Chromosomally Abnormal Persons Two main categories fall under this heading. The first, and most important, is the phenotypically normal person heterozygous for a balanced structural rearrangement (translocation, inversion, and insertion being the major forms). Second, there is the rare instance of persons who are themselves chromosomally unbalanced with either a full or a partial aneuploidy, and thus mostly phenotypically abnormal, and who present with questions of their reproductive potential. We will deal in detail with each situation in separate chapters, but we will consider the broad principles here.
2 Cytoplasm contains the mitochondria, and transmission of mitochondrial DNA is essentially maternal. The mitochondrial genome has been described, somewhat whimsically, as chromosome 25, or the M chromosome. In not otherwise referring to this “chromosome,” we are not seeking to deny its importance or interest! 3 Since, at the level of the chromatid, there are four elements, the word tetrad can also be used in this setting. In one sense, the cell at this stage of the cycle has 23 × 4 = 92 chromatids. At the molecular level, the number of single DNA strands is eight.
The Origins and Consequences of Chromosome Pathology •
29
(a) Replication
(b) Synapsis
Meiosis I
(c) Crossing-over
(d) Disjunction
(e) Meiosis II
Homolog of maternal origin
Homolog of paternal origin
FIGURE 3–2 Closer detail of chromosomal behavior during meiosis I. One crossover has occurred between the long arms of one chromatid of each homolog. In öogenesis, one of the two cells at (d) would be the first polar body.
BALANCED CARRIERS
In heterozygotes for some balanced rearrangements involving only small segments, the chromosomes may “ignore” the nonhomologous material they contain, and pair (this is “heterosynapsis”) and segregate much as would happen at a normal meiosis. In other balanced rearrangements, the inherent 30
•
BA SIC CONCEPTS
tendency to pairing dictates that homologous segments of rearranged chromosomes will align, as well as they are able to achieve this (“homosynapsis”). This may require the chromosome to be something of a contortionist, forming complex configurations such as multivalents and reversed loops. According to either scenario, the stage is set for the possibility of unbalanced segregation. The gametes produced—and
therefore the conceptuses that arise—are frequently unbalanced. In this context, a segmental aneuploidy is usually involved, that is, a part of a chromosome is present in the trisomic or monosomic state; or, rather frequently, a combination of trisomy for one segment and monosomy for another. Partial trisomy and partial monosomy are also referred to as duplication and deletion, respectively. In some rearrangements, recombination presents a further hazard. Inversions and insertions may produce a new recombinant (rec) chromosome that has a different genetic composition from that of the original rearrangement. A conceptus forming from a gamete containing it would inevitably be genetically unbalanced.
The cell division period is further subdivided into prometaphase → metaphase → anaphase → telophase. The chromosomes condense to enter prometaphase, and condensation continues into metaphase. Metaphase chromosomes align on the equatorial plate, and the spindle apparatus becomes attached to the centromere of each chromosome, consisting of its two kinetochores. Pulled at the kinetochores (centromeres), the chromatids of each chromosome then separate (disjoin) and are drawn in opposite directions (anaphase) and arrive at the opposite poles of the cell (telophase). Then the chromosomes decondense, the nuclear membrane reconstitutes, the cytoplasm constricts and divides, and two daughter cells now exist.
ANE UPLO ID IN D IV IDUA LS
Mosaicism due to Mitotic Error
In the individual who has a full aneuploidy, and in whom gametogenesis is able to proceed, in theory a trivalent may form, or a bivalent and an “independent” univalent. Either could lead, effectively, to a 2:1 segregation. This appears actually to be the case in trisomy 21; whereas in gonosomal4 states (XXX, XXY, and XYY) the “third” chromosome is, as it were, disposed of, and the great majority of gametes are normal. In the person with a partial aneuploidy due to an unbalanced rearranged chromosome, whether 46,(abn) or 47,+(abn), the abnormal chromosome may have an even (or near-even) chance to be transmitted in the gamete; but the opportunity to observe such outcomes rather infrequently arises.
A mitotic error can cause phenotypic abnormality by generating an abnormal cell line at some point during embryogenesis. If we focus on the end result, the feature distinguishing mitotic from meiotic errors is that the former typically produce a mosaic conceptus, while meiotic errors produce a nonmosaic abnormality. We define constitutional chromosomal mosaicism as the coexistence, within the one conceptus, of two or more distinct cell lines which are genetically identical except for the chromosomal difference between them, these cell lines having been established by the time that embryonic development is complete (the point at which the embryo becomes a fetus). Thus, the different cell lines are fixed in the individual and are a part of his or her chromosomal constitution. The earlier in embryogenesis that a mitotic error occurs, the greater the likelihood for a substantial fraction of the soma to be aneuploid, leading to increasing departure from normality of the phenotype. It is probable that many mitotically arising abnormalities lead to cell death, leaving no trace. Considering the enormous numbers of mitoses that proceed successfully, it is clear that the ordering of chromosomal disjunction during cell division must be a marvelously robust mechanism. A complex system of interacting components underlies the mechanism, including the cohesin multiprotein complex, among which are the similar RAD21 and REC8 proteins having crucial roles in mitosis and meiosis, respectively, and the synaptonemal complex
MITOSIS AND MOSAICISM The purpose of a mitotic cell division is faithfully to pass on an intact and complete copy of the parental cellular genome to the progeny cells. The mitotic cycle consists of the following sequence: gap-1 period (G1) → synthesis period (S) → gap-2 period (G2) → mitosis (cell division). The G1→S→G2 components together comprise the interphase period of the cell cycle. During the S period, the chromosomes replicate their DNA, thus converting from the single-chromatid to the double-chromatid state. Genetically active segments of chromosomes replicate earlier during the S period, while inactive segments, which include almost the entire inactivated X chromosome in the female, are late replicating.
4 Somewhat confusingly, some writers use this word to mean “gonadal-somatic,” when referring to mosaicism for a mendelian gene.
The Origins and Consequences of Chromosome Pathology •
31
proteins (Bardhan, 2010). Rare instances of marked mitotic instability indicate the existence of errors in the system, some at least of which may be genetically determined (Mikkelsen, 1966). For example, Miller et al. (1990) karyotyped a child because of major physical and neurodevelopmental defects, and he had cells trisomic and monosomic for almost every chromosome; only about a quarter were 46,XY. As a possible milder manifestation of this phenomenon, Fitzgerald et al. (1986) described a mother, herself physically normal, who had had three trisomic 21 conceptions. Her own tissues (blood, skin) were mostly 46,XX, but some cells had a variety of aneuploidies (47,+21, 47,+18, 47,XXX), indicating a proneness to chromosome malsegregation apparently operating both in meiosis and mitosis. A very early example of familial mosaicism, back in the days of solid-stain cytogenetics, concerns a family in which a mother and son were mosaic for a Robertsonian translocation, and the son had two mosaic children, a girl with Down syndrome who had four separate cell lines, and a boy with 45,X/46,XY mosaicism (Zellweger and Abbo, 1965). The more recently delineated mosaic variegated aneuploidy syndrome (p. 346) is now the classic example of a presumed “mitotic checkpoint mutation.” The quite common finding of loss of an X or a Y chromosome in an occasional cell in an older female or male population (and more notably in centenarians) may reflect “normal” age-related anaphase lag (Guttenbach et al., 1995; Bukvic et al., 2001; Russell et al., 2007). Aviv and Aviv (1998) refer to “age-dependent hidden mosaicism” and propose a role for the progressive shortening of telomeres in leading to somatic aneuploidies in older persons. Possibly, this chromosomal change might be an agent, rather than just a passive consequence, in the ageing process; a possibility the consideration of which is well beyond the remit of this book.
required a vast number of mitoses for their generation. The dozen cells checked in the laboratory are only a billionth of a percent of all the person’s cells, and we routinely (and, for practical purposes, not unreasonably) regard this minute fraction as a valid representative of the remaining 99.999999999%. Notwithstanding, we can surely suppose that one or more errors will have happened, during one or some of the many mitoses, and these will have produced a chromosomally abnormal cell line and the person is really a chromosomal mosaic. It seems plausible to imagine that unrecognized islands of mosaicism, involving a tiny number of cells—only a few thousand or a few dozen, perhaps—could well be a fairly frequent state. In fact, the cornea of the eye, and the brain, may show mosaic aneuploidy as an apparently normal phenomenon (Pettenati et al., 1997; Westra et al., 2008). Almost certainly, somewhere in their soma, everyone is a mosaic. DE T ECT I N G M OSA I CI SM
The great majority of cytogenetic studies are done on blood samples. Blood is a specialized tissue, and it may not necessarily reflect the karyotype elsewhere in the body. Skin fibroblasts are a more “basic” tissue, and skin biopsy has long been performed in the pursuit of a diagnosis of mosaicism. A particular case is that of the Pallister-Killian syndrome, due to 12p isochromosome (p. 305), which cannot be diagnosed on blood, and thus skin biopsy is a necessary procedure. Chorionic villi and amniocytes are the tissues assessed at prenatal diagnosis, and “confined placental mosaicism” is a well-recognized category. Other somatic tissues amenable to study, and thus allowing recognition of mosaicism, are the buccal mucosal cell and the urinary epithelial cell (Reddy and Mak, 2001; Stefanou et al., 2006). A somewhat different question is mosaicism in the preimplantation embryo (see the following section).
Mosaicism from the First Divisions of the Zygote A chromosome test on any normal person—a routine analysis from a sample of peripheral blood— would probably get a normal result (46,N). We would conclude from an analysis of a dozen or so cells from one specialized tissue that the rest of the soma is also 46,N. In most of the person’s tissue, this will be truly the case. But the body comprises a vast number of cells—a trillion (1012) or so—which 32
•
BA SIC CONCEPTS
The first few mitotic divisions from the one-cell zygote are particularly vulnerable to error, and this brief period of development needs to be considered separately. Insight into this vulnerability has come from experience in the in vitro fertilization (IVF) laboratory, with the application of preimplantation diagnosis. It may be that the early cleavage
pre-embryo has to rely on an inadequate supply of maternal cell-cycle control factors, conveyed in the egg, before being able to bring about its own autonomous production. Surprisingly large fractions of pre-embryos, on fluorescence in situ hybridization (FISH) analysis, are chromosomally mosaic. In one series, for example, in which 216 apparently normal IVF zygotes were followed through to the 2-4 cell and 5-8 cell stages, almost half were mosaic (Bielanska et al., 2002b). Going from the 2-4 cell to the 5-8 cell stage, the fraction rose from 15% to 50%. Often, the mosaicism was “chaotic,” that is, different cells had different aneuploidies. Pre-embryos that failed to advance had much higher levels of mosaicism than those whose development appeared to proceed smoothly, as naturally might have been expected. Munné et al. (2002b) studied over a thousand IVF embryos and deduced that a substantial fraction reflected mitotic nondisjunction, these embryos typically consisting of cells with normal, trisomic, and monosomic chromosomal constitutions. Interestingly, this category of embryo mosaicism was associated with increasing maternal age, possibly reflecting the decay of stored factors in the oöcyte as just mentioned. One must bear in mind that all these observations are made in the unnatural setting of in vitro development, and that the picture may be less abnormal in vivo. Further, FISH methodology may be less accurate than initially supposed, and mosaicism is observed much less frequently if SNP-microarray is the test procedure (Treff et al., 2010a). These matters are dealt with in some detail in Chapter 26. Insight into the timing of the abnormality can also be gained from inference in the study of mosaic individuals. Jacobs et al. (1997), in a study of Turner syndrome, observed that patients with Xq isochromosome mosaicism hardly ever have a 46,XX cell line: most are 45,X/46,X,i(Xq). This is what would be expected if the error happened at the very first mitosis of the initially 46,XX zygote. If it happened at the next two or three divisions, a 46,XX cell line would also have been present, 45,X/46,XX/ 46,X,i(Xq). If three cell lines are detected, an origin in a later mitosis can be assumed. For example, Stefanou et al. (2006) describe an abnormal infant with trisomy 20 mosaicism on blood, but with a
monosomic 20 cell line identified in urinary epithelial cells. The first division of the zygote may be especially, but certainly not uniquely, prone to error. CHI M ER I SM
Chimerism,5 which is to be distinguished from mosaicism, is the coexistence of more than one cell line in an individual, due to the union of two originally separate (“sibling”) conceptions. It could be imagined that twin blastocysts happen to make contact and then fuse (IVF may facilitate this happening), and this may be the more typical scenario. Alternatively, but likely very rarely, there might have been two sperm fertilizing an ovum and a polar body. A 46,XX + 46,XX chimera would most probably present as a normal female, whereas 46,XX + 46,XY could manifest an abnormality of sexual differentiation. An extraordinary example of chimerism is recorded in Wiley et al. (2002) of a malformed stillborn with 47,XY,+21 plus 47,XX,+12. The most usual form is “confined” chimerism, in which only one tissue—that is, blood—possesses the two cell lines. This is due to twin-to-twin (or feto-fetal) transfusion, when dizygous twins have intimately opposed placentae, allowing vascular connections (“anastomoses”) to form between them, with marrow colonization by the other twin’s hematogenous cells. Sudik et al. (2001), for example, describe a woman typing XY in 99% of peripheral lymphocytes on fluorescence in situ hybridization (FISH) analysis, but karyotyping 46,XX on three other tissues, including ovarian; she had had a twin brother, who had died as a neonate.6 TWINNING
Dizygous twinning is more frequent in mothers in their late thirties, and so it is not remarkable that occasionally twins are born, one with normal chromosomes, and the other with a maternal-age-related aneuploidy. Monozygous twinning could happen in an abnormal conception just as in a normal one, and the occasional instance of twins concordant for an abnormal karyotype is to be expected (Schlessel et al., 1990). Rather more remarkable is the case of monozygous twins discordant for karyotype—clearly
5 For the record, the chimera of classical mythology was “in the forepart a lion, in the hinder a serpent, and in the midst a goat.” 6 Somewhat stretching the analogy, Bianchi (2000) makes the intriguing suggestion that, due to the retention and persistence of fetal blood cells following delivery, every mother is, in a sense, a chimera (“microchimerism”).
The Origins and Consequences of Chromosome Pathology •
33
the adjective “identical” is inappropriate here! Rogers et al. (1982) studied monochorionic twin brothers, one 46,XY and the other 47,XY,+21 with Down syndrome, in whom marker analysis supported a diagnosis of monozygosity. The skin fibroblast karyotypes were nonmosaic, but both infants showed blood mosaicism, presumably from twin-twin transfusion in utero. In this type of twinning, the assumption is that either an initially 47,XY,+21 conceptus underwent splitting, with loss of a chromosome 21 then occurring in one of the newly created embryos; or, vice versa, a mitotic nondisjunction occurred in one monozygous embryo from an initially normal conception. Similarly, Nieuwint et al. (1999) describe two sets of monozygous twins, one of each pair being chromosomally normal and the other abnormal, monosomy X in one case and trisomy 21 in the other. Lewi et al. (2006) were able to assemble six such cases from one center, with cases of trisomy 21, trisomy 13, and monosomy X. A 47,XXY conception that led to monozygous twins of different gender, one 46,XX and the other 46,XX/46,XY, is reported in Zech et al. (2008). As for a structural rearrangement, Zeng et al. (2003) describe monozygous twins, one mosaic for a 1q duplication due to 46,X,der(Y) t(Y;1)(q12;q12), and the other twin normal; their analyses were consistent with a postzygotic error. Similarly, monozygous twin brothers, one 46,XY and the other with Xq isochromosome Klinefelter syndrome, 47,X,i(Xq),Y, are reported in Stemkens et al. (2007). Perhaps the most extraordinary circumstance of discordance in monozygous twins concerns the acardiac (that, is, lacking a heart) fetus. Trisomy 2 is one of the aneuploidies observed (Blaicher et al., 2000). An initially normal conceptus might generate a trisomy 2 cell line that then separates and produces the co-twin; or, an initially trisomic conceptus gives rise to a “corrected” lineage. It is only the presence of the normal twin that allows the acardiac co-twin to survive, with placental vascular connections providing blood circulation from normal to abnormal twin. We have seen such a case due to trisomy 3, with the affected acephalic, acardiac fetus of barely recognizable human form. Somatic Recombination in Homologs. Genetic exchange can take place, as a normal event, during a
mitotic cycle, either involving the pair of homologous chromosomes, or the sister chromatids of one chromosome. The cytogenetic demonstration of sister chromatid exchange (SCE) is rather dramatic (Fig. 21–2). Should the SCE be unequal, tandem duplication and deletion lines may be generated. If the deletion line is lost, a normal/duplication mosaicism results (Rauen et al., 2001). According to the somatic extent of the abnormal cell line, the phenotype may or may not be affected; and according to its involvement in the gonad, a reproductive risk may or may not apply.
NONDISJUNCTION Nondisjunction in Meiosis C LASS I CA L D ES CR I PT I O N
Nondisjunction is remarkably frequent, and in consequence many human conceptions, perhaps about a quarter, are trisomic or monosomic. Nondisjunction is defined as the failure of homologous chromosomes to segregate symmetrically at cell division. The classical description of the mechanism of meiotic nondisjunction is as follows. In a chromosomally normal person, if the pair of homologs comprising a bivalent at meiosis I fail to separate (fail to disjoin7), one daughter cell will have two of the chromosomes and the other will have none. This is 2:0 segregation (Figs. 3–3a and 3–4, upper). In other words, one cell is disomic for that homolog, and the other is nullisomic. Nondisjunction may occur in meiosis II, meiosis I having proceeded normally. In meiosis II, it is the chromatids that fail to separate (Fig. 3–3b). Following these nondisjunctional errors, the conceptus, at fertilization, ends up trisomic or monosomic, assuming the other gamete to be normal (Fig. 3–5a,b). Trisomy or monosomy in the offspring of normal parents is called primary trisomy or primary monosomy. Most nondisjunction, at least in oögenesis, is assumed to occur at the first meiotic division. This is the conclusion reached with the most extensively studied chromosomes, nos. 8, 13, 15, 16, 18, and 21 (Nicolaidis and Petersen, 1998). Indeed, in the case of trisomy 16, every case is proposed to be due to a maternal meiosis I error. With respect to the X chromosome, about 90% of nondisjunctions leading to
7 Note that disjunction is a normal process, and nondisjunction is not; there is no such word as dysjunction.
34
•
BA SIC CONCEPTS
(a) Nondisjunction at meiosis I
NONDISJUNCTION
Meiosis I
Meiosis II
Disomic gametes
Nullisomic gametes
(b) Nondisjunction at meiosis II
Meiosis I
Meiosis II
NONDISJUNCTION
Disomic gamete
Nullisomic gamete
Normal gametes
FIGURE 3–3 The classical view of the mechanics of nondisjunction. The asterisked gamete reflects the complement of the oöcyte in Figure 3–4 (upper). In öogenesis, one of the two cells following meiosis I would be the first polar body, which might or might not proceed to meiosis II.
the 47,XXX state are of maternal origin. At least half of the maternal X nondisjunctions that cause 47,XXX and 47,XXY arise from meiosis I errors, and about a third in meiosis II (Thomas et al., 2001). Meiosis II is the site of some autosomal nondisjunctions, and in fact most trisomy 18 may result from
this stage. Autosomal nondisjunction in spermatogenesis results in far fewer abnormal conceptions than from oögenesis, with the 22 autosomes mostly having about an equal likelihood to undergo nondisjunction, although some (chromosomes 16, 21, 22) appear to be more vulnerable (Shi and Martin, 2000b).
The Origins and Consequences of Chromosome Pathology •
35
FIGURE 3–4 Oöcytes at metaphase of meiosis II, showing nondisjunction of a G-group chromosome having occurred at the preceding first meiotic division. Upper, oöcyte with classical nondisjunctional disomy, showing an additional G-group double-chromatid chromosome. Possibly the arrowed pair are chromosome 21s, and the karyotype 24,X,+21. Lower, oöcyte with “predivisional” disomy, showing an additional G-group single chromatid. The arrowed pair may be chromosome 21s, and the karyotype 24,X,+21cht. (From Kamiguchi et al., 1993, courtesy Y. Kamiguchi.)
36
•
BA SIC CONCEPTS
Only in trisomy 2 among the autosomes is there a substantial paternal contribution, with close to half reflecting a meiotic error in spermatogenesis (Hassold, 1998; Robinson et al., 1999). Given the frequency with which nondisjunction happens, it is not at all surprising that instances of double trisomy are known. The reader with a sense of history will want to review the 48,XXY,+21 case described in Ford et al. (1959); and a very few other cases, proceeding through to live birth, have followed, the most common combination being trisomy 21 along with an additional sex chromosome (Li et al., 2004; Tennakoon et al., 2008). The observation is more frequently made, and double autosomal combinations seen, in the analysis of products of conception from spontaneous abortions (Micale et al., 2010). Through the IVF laboratory, a very much larger number have come to light, and often with several coexisting aneuploidies; for the most part, these embryos would not even proceed through to the stage of implantation. While autosomal aneuploidy overwhelmingly has its origin in oögenesis (and meiosis I at that), male gametogenesis has a major part to play in the generation of sex chromosome abnormalities. Obviously enough, the 47,XYY state, when it is due to a meiotic error, must be due to paternal nondisjunction (at meiosis II, logically), with the production of a 24,YY sperm. Some may represent a postzygotic error (Robinson and Jacobs, 1999). As much as half of 47,XXY Klinefelter syndrome is due to nondisjunction having occurred in spermatogenesis, giving a 24,XY sperm. X-Y nondisjunction is predisposed following an absence of recombination in the primary pseudoautosomal regions (PARs) of the X and Y at meiosis I. And monosomy X is mostly due to absence of a paternally contributed sex chromosome. Sequential nondisjunctions at both meiotic divisions could lead to tetrasomy, and this is the basis of some X chromosomal polysomy (Hassold et al., 1990b; Deng et al., 1991). Simultaneous nondisjunctions of two chromosome pairs can lead to double aneuploidy, as noted earlier. Simultaneous parental nondisjunctions, both gametes being disomic, is rare, but not unknown, and is another route to double aneuploidy, and for example Robinson et al. (2001) describe 48,+14[pat],+21[mat] in a spontaneous abortion. If one gamete is disomic and the other nullisomic, for the
(a)
Disomic gamete
Normal gamete
Trisomic conceptus
Nullisomic gamete
Normal gamete
Monosomic conceptus
Disomic gamete
Nullisomic gamete
Uniparental disomy
(b)
(c)
FIGURE 3–5 Aneuploid gametes producing an aneuploid conceptus (a and b), and aneuploid gametes producing uniparental disomy (c).
same chromosome, one parent has contributed both members of the homologous pair, and the other none (Fig. 3–5c). This is uniparental disomy due to “gametic complementation,” an event of extreme rarity. Simultaneous errors of nondisjunction and other rearrangement would typically be quite coincidental, such as a child having both XXY Klinefelter syndrome (maternal nondisjunction), and del(15)(q11.2q13) Prader-Willi syndrome (paternal deletion) (Nowaczyk et al., 2004). Complete nondisjunction is an expression that could be applied in the case of triploidy, when this is due to the retention of the polar body within the ovum (Martin et al., 1991).
T HE CH R O M AT I D “ PR ED I V I S I O N ” HY POT H ES I S O F A N G EL L
An alternative mechanism for nondisjunction is based on the premise that precocious separation of chromatids (“predivision”) during meiosis I is the crucial factor. This is Angell’s (1997) hypothesis, and it is based upon direct oöcyte observations. Three sequential events comprise the gist of this theory (Fig. 3–6). First, the (double-chromatid) homologs fail to pair during meiosis I8; or, if they do pair, they separate again before meiosis I is complete. In other words, instead of the two (doublechromatid) chromosomes existing as a conjoined bivalent, they exist as two separate univalents. Second, these univalents are prone to “predivide”—that is,
8 If the homologs had never joined together, then they could not disjoin. In that sense, nonconjugation might be a more accurate word than nondisjunction, a point Sturtevant and Beadle (1962) made many years ago. Following Angell, predivision may be a proper word. Nonetheless, nondisjunction is well entrenched in the genetic lexicon, and its general meaning of “the inclusion of both daughter chromosomes in the same nucleus, by whatever mechanism” (Miller and Therman, 2001) is well accepted.
The Origins and Consequences of Chromosome Pathology •
37
the separation of the two chromatids that should normally happen at meiosis II instead takes place while they are still in the first meiotic cycle. This could happen to both univalents or just the one; and these would then exist as single-chromatid chromosomes. Third, at anaphase of meiosis I, these doubleor single-chromatid chromosomes segregate to the oöcyte and polar body independently. The oöcyte in Figure 3–4 (lower) may be an example of asymmetric segregation due to this process, having received a double-chromatid and a single-chromatid chromosome. Sandalinas et al. (2002) provide some corroborative support for Angell’s hypothesis in their findings on direct analysis of fresh oöcytes, with both predivision and nondisjunction more frequent
in the gametes of women over age 35 years than under 35, and more often observed in the smaller chromosomes. Similarly, Pellestor et al. (2005) and Treff et al. (2009a) show, using oöcytes obtained during IVF procedures, that predivision is a more frequent cause of aneuploidy than is classical nondisjunction, and furthermore is related to increasing maternal age. The phenomenon may not be confined to the female: Uroz et al. (2008) observed premature chromatid separation in sperm of an infertile man. Certain terminologies and nomenclature may be mentioned here. A gamete with an extra chromosome is hyperhaploid, with a karyotype
No synapsis
Predivision
RANDOM SEGREGATION
Meiosis I
Meiosis II
Disomic gamete
Normal gametes
Nullisomic gamete
FIGURE 3–6 Nondisjunction following “predivision” of one homolog into its component chromatids in meiosis I (Angell’s hypothesis). The asterisked gamete reflects the complement of the oöcyte in Figure 3–4 (lower). In öogenesis, one of the two cells following meiosis I would be the first polar body, which might or might not proceed to meiosis II. 38
•
BA SIC CONCEPTS
written, say, 24,X,+21. A gamete missing a chromosome is hypohaploid. (e.g., 22,Y,–21). If, at meiosis I, the extra chromosome is present only as a single chromatid (e.g., the asterisked oöcyte in Fig. 3–6), the abbreviation cht is used: thus, 24,X,+21cht. In addition, the ISCN provides nomenclature for meiotic cells, and an extra 21 at meiosis I, present as a univalent, would be denoted as MI,24,+I(21) (ISCN 2009). CAUSES O F N O N D ISJU N CT I ON
The great majority of aneuploidy due to nondisjunction arises in oögenesis. A particular vulnerability of maternal meiosis may lie in the degradation, over time, of factors that underpin the adhesion of the homologous chromatids of the bivalent. This failure of snug apposition, then, leads the chromosomes to adopt unstable positions when meiosis resumes. The particular unstable position will depend, where there is just one chiasma, upon whether its site is toward the middle or toward one end of the chromosome. This can then allow the pairs of chromatids, only loosely attached to each other at this single chiasma, to act as independent univalents at the first meiotic division (Wolstenholme and Angell, 2000; Pellestor et al., 2002; Yuan et al., 2002). One apparently rare mendelian basis is the SYCP3 gene, coding for one of the synaptonemal complex proteins, mutation in which affects meiosis both in the female, to produce aneuploid oöcytes, and in the male, to cause spermatogenic arrest (Bolor et al., 2009). Another theory has it that the motor proteins associated with the centromere may be a point of vulnerability. Chromosomes move along the spindle to their appropriate destinations by the active intervention of these motor proteins, and if they are not working properly, chromosomes may end up being not where they ought to be (Hodges et al., 2002). Quality checking, which is stringently applied in the male, is poorly effective in the female, and so the maturing of an aneuploid oöcyte is not prevented; as Hunt and Hassold (2002) suggest, Nature seems to have erred in putting less protective investment into the more scarce gamete. While these meiosis-control factors may be the proximate cause of failed disjunction, what background attributes might lead to a loss in its integrity? Of course, advanced childbearing age is an obvious answer. A very telling insight comes from the work of Battaglia et al. (1996). These investigators
sampled oöcytes at meiosis II metaphase from younger (20–25 years) and older (40–45) volunteers who were having normal menstrual cycles. They did not look at individual chromosomes but rather at the disposition of the spindle and the metaphase chromosomes as a whole. They made the most striking findings according to the ages of the women: a symmetrical and neatly arrayed complex was seen in the younger women, while in the older women the spindle was askew and the chromosomes a-jumble, as shown in Color Figure 3–7 (see separate color insert). It is not difficult to accept that this structural disorganization would undermine the ability of the chromosomes of the oöcyte then to undergo regular segregation. And in a paper that could have been subtitled “A Tribute to the Polar Body,” Fragouli et al. (2011) show how revealing can be an analysis of the chromosomes of this discarded remnant. Exploiting the fact that the polar body’s karyotype is, in a sense, the opposite of that of the oöcyte, and that the first and second polar bodies reflect segregations at meiosis I and II, respectively (Fig. 3–1), they could show that maternal age has a very strong association both with nondisjunction and with predivision of chromatids, and that meiosis II becomes, with age, the more vulnerable division. Accumulation of mitochondrial DNA mutations might be a further factor causing compromise of the functioning of the support structures underpinning the meiotic mechanisms (Schon et al., 2000), and Seifer et al. (2002) demonstrated an increasing frequency with age of a particular mtDNA deletion, which is otherwise known to be age related, in the cells that surround the oöcyte within the follicle (the granulosa cells). Thus, support may be drawn for the view that a gradual deterioration in the integrity of the meiotic apparatus with increasing age of the mother is the major factor in predisposing to meiotic nondisjunction in female gametogenesis. Individual variation may mean that some women are more prone to these effects than others (Nikolaou and Templeton, 2004). Not that the young are immune. Fragouli et al. (2006a), in a paper dedicated to the memory of the 18-year-old patient whom they had studied, analyzed oöcytes harvested ahead of her chemotherapy for a marrow malignancy which, had she lived, might have enabled fertility. Of 11 oöcytes and 7 first polar bodies able to be analyzed, one egg had a singlechromatid X and could have gone on to a monosomy X conception, while another egg was inferred
The Origins and Consequences of Chromosome Pathology •
39
(a)
22 year old
24 year old
42 year old
(b)
40 year old
FIGURE 3–7 Meiosis II oöcytes from younger and older women, illustrating what may be the physical basis of the maternal age effect. The microtubules of the spindle stain green, and the chromosomes stain orange. The tracing identifies these components, and the smooth or wavy lines suggest, respectively, an intact or a degenerating spindle apparatus (the ages of the women indicated). The chromosomes are well organized at the metaphase plate at the equator of the cells in the younger women (the 22-year-old’s oöcyte, on the upper left, is viewed on a tilt). In contrast, the 40-year-old’s oöcyte shows the chromosomes in disarray. The 42-year-old woman’s oöcyte has one chromosome, at the top, dislocated from the metaphase plate, and the disposition of the other chromosomes at the equator is not as regular as in the younger women. (The color photographs are from D. E. Battaglia et al., (1996), Influence of maternal age on meiotic spindle assembly in oöcytes from naturally cycling women, Human Reproduction 11:2217–2222. Courtesy D. E. Battaglia; reproduced with the permission of Oxford University Press and Human Reproduction.) See also separate color insert.
(via its polar body) to have an additional X and 21 chromatid, and the conception could have been 48,XXX,+21. The introductory sentence of this paper is worth quoting: “Humans as a species are not as fertile as other mammals”; and, as already noted, it is at meiosis in the oöcyte that much of this susceptibility resides.
Nondisjunction in Mitosis NORM A L Z YG OTE
Mitotic (somatic, postzygotic) nondisjunction is the major mechanism in the causation of mosaicism. Nondisjunction can occur in an initially normal (46,N) zygote, with the generation of mosaicism for a trisomic and a concomitant monosomic line, as well as the normal line (Fig. 3–8a). In autosomal nondisjunction, growth of the monosomic cell line is severely disadvantaged, and it will very probably die out in early embryogenesis, leaving just the normal and the trisomic cell lines comprising the individual.9 Mosaic Down syndrome, with the karyotype 46,N/47,+21, is the classic example. In one particular autosomal aneuploidy, trisomy 8 mosaicism, somatic nondisjunction accounts for the great majority of cases (Karadima et al., 1998). Actually, about 5% of standard apparently nonmosaic 47,+21 is also due to a mitotic defect (Antonarakis et al., 1993), with the “third” chromosome 21 equally likely to be maternal or paternal. In 3% of apparently nonmosaic 47,XXY and 9% of 47,XXX, the error was postzygotic, presumably prior to the formation of the inner cell mass (MacDonald et al., 1994). As noted earlier, the nature of the mosaicism can indicate the likely time of its generation. An aberrant mitosis involving the X chromosome, in an initially 46,XX zygote, may generate X and XXX cell lines, both of which would be survivable. If this happens at the first mitosis, X/ XXX mosaicism would result. If at any later mitosis, a normal cell line would exist, and the mosaic state would be X/XX/XXX (Fig. 3–8b). The same can happen in a 46,XY zygote, with an X/XYY, or an X/ XY/XYY mosaicism resulting (the gender in the embryo being determined according to the sex chromosome composition of gonadal tissue). More than
FIGURE 3–8 Generation of mosaicism. (a) Postzygotic nondisjunction in an initially normal conceptus. In this example, one cell line (monosomic 21) is subsequently lost, with the final karyotype 46,N/47,+21. (b) Postzygotic nondisjunction in an initially normal 46,XX conceptus, resulting in 45,X/46,XX/47,XXX mosaicism. (c) Postzygotic anaphase lag in an initially abnormal 47,+21 conceptus; this leads to a “corrected,” or “rescued,” normal cell line.
9 A very rare example of autosomal monosomy/disomy/trisomy mosaicism was identified in the abnormal baby reported in Stefanou et al. (2006), mentioned earlier. Only 1 cell in 200 on blood showed 47,XY,+20, and disomy demonstrated on buccal mucosal FISH and skin fibroblast analysis, but 39/50 cells from urinary sediment were monosomic.
The Origins and Consequences of Chromosome Pathology •
41
one mitotic error can happen, separate in time and place; for example, DeBrasi et al. (1995) identified concomitant 45,X and 47,XX,+8 (and 46,X,+8) in a woman with clinical features of both trisomy 8 and Turner syndrome, in whom the molecular study supported the hypothesis of an originally 46,XX conception. Two sequential abnormal events can lead to the intriguing situation of nonidentical monozygous twins of opposite sex. The mitotic loss of a Y chromosome in a 46,XY conceptus could produce X/XY mosaicism; and then a splitting of the conceptus could produce monozygous twins in which the distribution of the X and XY lineages might differ. Costa et al. (1998) studied two sets of X/XY mosaic twins, each set comprising a girl and a boy. In one set, the girl was diagnosed clinically as Turner syndrome and had a 45,X[23%]/ 46,XY[77%] karyotype on blood analysis, while her male co-twin was 45,X[16%]/ 46,XY[84%]. In the other set, the girl had mixed gonadal dysgenesis, and her karyotype was 45,X[24%]/46,XY[76%]. Her brother’s karyotype, on blood, was 45,X/46,XY with the same ratios as his sister; but on fibroblast culture he was nonmosaic 46,XY. He had absence of one testis. Fetal blood mixing, due to vascular connections in the shared placenta, may have contributed to his 45,X cell line. A most extraordinary suggestion, and one which would overturn much of the received wisdom about the generation of trisomy, comes from Hultén10 et al. (2008, 2010), who studied ovarian tissue from apparently normal female fetuses, and observed, in eight out of eight cases, and with a total of 12,634 cells analyzed, a very low-level trisomy 21 mosaicism: 0.5%, with a range of 0.2%–0.9%. In contrast, no such mosaicism existed in the testes of male fetuses. They speculate that, in an initially 46,XX embryo, precursor gametic cells may be vulnerable to mitotic error; and they reason from this observation that “most women may be trisomy 21 ovarian mosaics.” This they propose as the basis for the predominance of a maternal origin of the extra chromosome in Down syndrome and other aneuploidies. The age effect may be due to a different maturity of
the trisomic oöcyte. This will be an interesting debate to follow. A NEUPLO I D ZYG OT E
Nondisjunction can occur in a postzygotic mitosis in a conceptus that is initially trisomic for an autosome (say, 47,+21). Thus, one copy of the homolog in question is lost. The same result may be due to the mechanism of anaphase lag. (In this latter case, the chromosome fails to connect to the spindle apparatus, or is tardily drawn to its pole, and fails to be included in the reforming nuclear membrane. On its own in the cytoplasm, it will form a micronucleus and soon be lost.) This converts the trisomy in this cell to 46,N. Its descendant cells are 46,N, and the karyotype of the conceptus is, say, 46,N/47,+21 (Fig. 3–8c). Most mosaic trisomy/disomy 13, 18, 21, and X arises in this way, for example, 47,XXY → 46,XY/47,XXY (Robinson et al., 1995). A conceptus with what might be called “interchange tertiary trisomy”—that is, a 47-chromosome count, with the two translocation chromosomes and an additional copy of one of the derivative chromosomes—might generate a cell line with the balanced state, if one of the derivatives is lost postzygotically. Thus, a zygote with, for example, a 47,t(1;2),+der(1) karyotype might acquire a cell line with 46,t(1;2). If this cell line included bloodforming tissue, but if much of the soma otherwise consisted of cells with the unbalanced state, a phenotypically abnormal child could have, on blood sampling, a balanced translocation karyotype. Such a case is presented in Dufke et al. (2001); and speculatively, this scenario might be a rare contributor to the apparent slight excess of abnormal children among the balanced carrier offspring of translocation carrier parents (p. 109). Postzygotic “Correction” of Aneuploidy and Uniparental Disomy. If the conversion of trisomy to disomy—sometimes referred to as “correction” or “rescue”—occurs prior to the formation of the preembryo, and if the 46,N line then gives rise to the pre-embryo, the embryo will be nonmosaic 46,N. According to which one of the three chromosomes was lost, normal biparental disomy in the embryo could be restored, or uniparental disomy (UPD) could result (Fig. 3–9). This is a much more common
10 Dr. Maj Hultén was the second person in the world to know that the human chromosome number was 46 (Harper, 2006).
42
•
BA SIC CONCEPTS
Abnormal gametogenesis in one parent
Meiosis I
Normal gamete from other patent
Meiosis II
Conception
Zygote
Pre-embryo
FIGURE 3–9 Uniparental disomy from “correction” of a trisomic conceptus by loss of a homolog. Nondisjunction* at meiosis I, followed by postzygotic loss** of one homolog, causes uniparental heterodisomy. (If, for example, this were chromosome 15, and the meiotic nondisjunction occurred in the mother, the child would have Prader-Willi syndrome.) Nondisjunction at meiosis II would cause uniparental isodisomy.
mechanism of UPD than the scenario of coincidental nondisjunctions in the genesis of both gametes as depicted in Figure 3–5c. It is at prenatal diagnosis, typically, that the fact of this rescue mechanism comes to be discovered, with trisomy seen at chorionic villus sampling (CVS), and disomy at a subsequent amniocentesis (Sirchia et al., 1998). Chromosome 15 is of particular concern, and Purvis-Smith et al. (1992) and Cassidy et al. (1992)
provide historic illustrations in pregnancies showing 47,+15 at CVS, with conversion to 46,N at amniocentesis; the infants had upd(15) mat, which produces the phenotype of Prader-Willi syndrome. Walczak et al. (2000) showed the same thing retrospectively, in demonstrating trisomy 15 by FISH on archived placental tissue. An inference of “rescue” may be made in the case of UPD discovered because of isozygosity for a
The Origins and Consequences of Chromosome Pathology •
43
recessive gene, and an example of this is deafness due to the connexin-26 gene. Yan et al. (2007) report a child presenting with deafness due to homozygosity for the 35delG mutation, for which his father, but not his mother, was a carrier. As it transpired, the child had upd13(pat), with isodisomic and heterodisomic segments of chromosome 13, the segment in 13q12.1, which contains the connexin-26 locus, being one of the regions of isodisomy. Quite possibly, this had been a trisomic 13 conception but rescued due to discarding one of the chromosomes, which happened to be the maternal chromosome 13. Had it not been for the coincidence of the father’s heterozygosity for the 35delG mutation, the rescue would have been entirely successful. Postzygotic correction can also happen in the other direction, as it were: to convert a monosomic zygote into a disomic one. It is rarely recognized. Quan et al. (1997) report a girl, 46,XX, with Duchenne muscular dystrophy due to a homozygous deletion of exon 50 of the dystrophin gene. She had homozygosity of the X chromosome for all of the tested marker loci: apparently, a complete maternal uniparental isodisomy X. Even a meiosis II nondisjunction would probably have had some heterozygosity, due to recombination at meiosis I; and so Quan et al. propose a mitotic mechanism. A 45,XO conception, from a 22,O sperm + 23,X egg at syngamy, underwent duplication, or possibly nondisjunction, of the single X chromosome. Unfortunately, this X chromosome carried a de novo Duchenne mutation. It remains an open question how many monosomy X conceptions, the X chromosome being normal, are successfully corrected to produce normal 46,XX,upd females (Schinzel et al., 1993).
STRUCTURAL REARRANGEMENT The following classical structural rearrangements may be listed: translocations, insertions, inversions, isochromosomes, duplications, deletions, and complex rearrangements. Smaller rearrangements, which may be referred to as “genomic rearrangements,” are of a size discernible on high-resolution banding (3–5 Mb), or may be of submicroscopic size (typically measured in kilobases) (Gu et al., 2008). All arose de novo at one point: whether with the index case in whom the abnormality was discovered, or in a parent or more distant ancestor, with a balanced (or, infrequently, unbalanced) form transmitted 44
•
BA SIC CONCEPTS
thereafter in the family. Jacobs (1981) has derived the following mutation rates for the generation of de novo classical rearrangements: 1.6 × 10–4 per gamete for the balanced reciprocal translocation, and 2.9 × 10–4 per gamete for unbalanced rearrangements (a mutation rate that is magnitudes greater than those typically seen in autosomal dominant conditions). The illegitimate breakage and reunion that produces these rearrangements is typically due to the apposition of chromosomal segments containing DNA sequences with a high degree of homology— “paralogous sequences,” also known as “duplicons.” Palindromic AT-rich sequences comprise the basis of “hot-spots” at 11q23, 17q11, and 22q11, leading to recurrent translocations t(11;22) and t(17;22) arising with breakpoints at these sites (Kurahashi et al., 2010). Most breakpoints are in nontranscribed DNA, and thus, for the most part, contribute no untoward effect upon the phenotype. If the breakpoint in one chromosome were to disrupt a gene, phenotypic defect is the likely consequence; the Developmental Genome Anatomy Project (Higgins et al., 2008) aims to collect such cases, in order to understand the precise nature of the molecular perturbation due to the breakpoint, and the process by which the phenotypic defect arises. We discuss possible mechanisms of formation in more detail in the appropriate chapters.
Setting in Which Rearrangement Occurs Mutations causing chromosomal rearrangement can occur during both meiosis and mitosis, and in the gonad of either sex. But (in contrast to the maternal risk for whole-chromosome aneuploidy) it is the male who is the susceptible parent, at least with respect to unique (nonrecurrent) rearrangements (Shaffer and Lupski, 2000; Kurahashi et al., 2010; Thomas et al., 2010). Balanced nonrecurrent de novo rearrangements show a paternal age effect, and likely arise at a premeiotic mitosis, whereas meiosis is the site of generation of unbalanced nonrecurrent de novo rearrangements, with no age effect discerned. Only in recurrent rearrangements, such as the Robertsonian translocation, is a maternal bias seen. A different view is proposed by Vanneste et al. (2009), who suggest that chromosomal breakagefusion-bridge cycles in the cleavage stage (day 1–3) embryo may be a frequent mechanism for several types of rearrangement, including mosaics.
Balanced and Unbalanced Rearrangements Structural rearrangements can be balanced, with the correct amount of genetic material in a cell, or unbalanced, with a deletion and/or duplication of genetic material. Arguing somewhat circularly, in the phenotypically normal person it is inferred that, although such an individual’s genetic material is in a different chromosomal arrangement, it is present in the correct (balanced) amount and functioning properly. It is irrelevant to the person’s health, other than his or her reproductive health. It may be helpful in explaining this to think of the person’s genome as a recipe book—a series of instructions for everything that is genetically determined. If an error occurs in the pagination (a translocation), and, say, pages 17 to 24 are inserted between pages 36 and 37, the recipes are all still there; they are still perfectly capable of being read. If a sequence of pages is inserted upside down (an inversion), one need only turn the book around to read them. At the cytogenetic level, one can only use the term “apparently balanced.” A not infrequent clinical and laboratory puzzle is the instance of a phenotypically abnormal individual who has an apparently balanced rearrangement. Might it be, at a submicroscopic level, that genetic imbalance really exists? Even the highest-resolution banding does not reveal a genetic imbalance at the level of a few kilobases of DNA; but now with microarray methodology, light is being cast on some of these cases, with a substantial fraction revealed as being, in fact, unbalanced (Schluth-Bolard et al., 2009; Vandeweyer and Kooy, 2009), or “molecularly balanced” but disrupting a gene that could plausibly be pathogenic (Cacciagli et al., 2010). If the (normal) parent has the apparently identical chromosome, the problem becomes more complex. These matters are dealt with in more detail in Chapter 5. A rearrangement that is balanced at the genomic level may yet lead to a phenotypic consequence due to “position effect.” Brown et al. (2010) review examples and describe their own case of a family with a segregating paracentric inversion, inv(7) (q21.3q35). The proximal breakpoint at 7q21.3, and which was associated with a 5 kb deletion, led to loss of an enhancer of the genes DLX5 and DLX6, and
also caused these genes to become functionally separated from other presumed regulatory elements. As a result, the activities of DLX5 and DLX6 were compromised; and since these genes influence development of the inner ear and aspects of craniofacial and limb formation, deafness and facial and limb malformation resulted. This may be considered an “epigenetic” effect, and we now go on to discuss this concept.
EPIGENETICS AND GENOMIC IMPRINTING A formal definition of an epigenetic effect includes these points: the DNA sequence of a particular gene remains unaltered, but the ability of this gene to be expressed is altered. The expression “genomic imprinting” is applied in the setting of epigenetic effects that are applied during germline transmission. Some parts of some chromosomes are subject to genomic imprinting as a normal occurrence, and this imprinting is parent specific: that is, genes in the chromosomal segment are expressed, or not expressed, according to whether the chromosome had been transmitted in the sperm or in the ovum (“parent-of-origin effect”). An imprinted segment takes up an “epigenetic mark,” and the gene or genes in this segment are not expressed, leaving it to the corresponding locus or loci on the homologous chromosome from the other parent to be the only source of expression. Imprinting can therefore lead to a differential activation status of the two alleles of the locus or loci concerned: one of the pair is functional, and the other is “silent.” When the phenomenon was first appreciated in humans, it was naturally suspected that many forms of congenital abnormality might be due to aberrant imprinting. As it has transpired, however, the practical application of genomic imprinting appears to be confined to a rather small number of cytogenetic conditions. Nevertheless, the theoretical interest is considerable. Most of the autosomal genome is not subject to imprinting, and it is functionally disomic. That is, with each locus having a pair of alleles, each of the pair is functionally active, contributing more or less equally to the genetic output from that locus.11 This is biallelic gene expression. A minority of the genome is subject to imprinting and requires only one of
11 But exceptions exist, and around 5% of autosomal genes are randomly expressed from only one or other parental allele (Gimelbrant et al., 2007).
The Origins and Consequences of Chromosome Pathology •
45
the pair of alleles to be active, while the other one becomes inactivated; in other words, the locus is functionally monosomic, with a genetic output from only one allele. This is monoallelic expression. If the allele of maternal origin is inactivated, only the allele of paternal origin is functionally active; and vice versa. Following conception, the imprint remains through cycles of postconceptional somatic mitoses: the chromosome “remembers” the sex of the parent who contributed it (put differently, it retains its epigenetic mark). The imprinting pattern may be specific to a certain tissue, or to a certain developmental stage (Ideraabdullah et al., 2008). Thus, in some tissues a gene may express monoallelically, while in other tissues biallelic expression is retained; or a gene may express monoallelically in a specific tissue at one stage in embryogenesis, and biallelically thereafter. X chromosome inactivation is a special case. Parent-of-origin imprinting is a normal mechanism of gene regulation. It is mediated through a process taking place during gametogenesis, of which the physical basis includes methylation of cytosine bases within the gene(s), or in controlling sequences upstream of it. This process is reversible, and in the “life” of an autosomal allele or chromosomal segment, as it passes from individual to individual down the generations and across the centuries, imprinting— the epigenetic mark—will be acquired, maintained, lost (“erased”), reacquired (“reset”), and lost again according to the sexes of the individual through whom it is transmitted. Throughout, it retains the same DNA sequence.
Mechanisms Whereby Functional Genetic Abnormality Can Arise In the context of imprinting, we may consider three categories of functional genetic defect. These are as follows: uniparental disomy with overexpression and/or nonexpression of genes in certain chromosomal segments; deletion with nonexpression; and relaxation of imprinting with overexpression. (1) Uniparental disomy will lead to either biallelic expression, or no expression, at the locus or loci within the imprintable segment. (2) If a deletion removes a chromosomal segment that would otherwise have been “silenced,” all that is lost is a nonfunctioning genetic segment, and there is no untoward consequence. On the other hand, if the deletion removes the segment on the active chromosome, the corresponding part of the other 46
•
BA SIC CONCEPTS
homolog is inactive, and so neither chromosome will be genetically functioning in this segment; in a sense, the silent allele is unmasked. (3) Relaxation of imprinting allows a segment that should be nonexpressed to lose its imprint. The locus or loci contained therein will be operating biallelically, which will be, in this case, at double normal capacity. These mechanisms are dealt with in detail in Chapter 22. Another category of epigenetic effect is that imposed by compromise of controller elements, such that the client gene, which is of itself normal, is inappropriately nonfunctioning; the “position effect” due to a translocation separating the controller and the client has been mentioned earlier.
TRINUCLEOTIDE-REPEAT TRANSMISSION Every counselor will be familiar with the concept of the trinucleotide expansion disorders. The first to have been recognized as such, in 1991, was Kennedy spinobulbar muscular atrophy, followed shortly by the fragile X syndrome. With the fragile X syndrome, the number of trinucleotide repeats can change as the allele is transmitted from parent to child. Having reached a critical number, the gene’s function is compromised, and phenotypic abnormality declares itself. The rare folate-sensitive fragile sites are most likely due to expansions of naturally occurring highly polymorphic CCG sequences (Sutherland, 2003).
CONSEQUENCES OF GENETIC ABNORMALITY STRUCTURAL IMBALANCE Chromosome imbalances are harmful because of the fundamental reason that some (not all) genes are dosage sensitive. In duplications, there is 150% of the normal amount of this chromosomal segment; and in the deletion, 50% of the normal amount. The imbalance involves a whole chromosome (full aneuploidy), or a part of a chromosome (partial aneuploidy, segmental aneusomy). An incorrect amount of dosage-sensitive genetic material in every cell of the conceptus distorts its development to a greater or lesser extent. Large losses or gains almost invariably set early development so awry that abortion occurs. Lesser imbalances may be compatible with continued intrauterine survival, but with the
eventual production of a phenotypically abnormal child. Very minor imbalances may cause defects that are not readily detectable in early infancy; and some chromosomal “defects” may be without phenotypic effect. However, as a first principle, anything but 100% of the normal amount of (at least autosomal) genetic material produces a less than 100% normal phenotype. Mental defect is the almost universal consequence of autosomal imbalance, and vice versa, much mental defect is due to a chromosome abnormality. It is generally too simplistic to think of deletions and duplications leading to opposite qualities of phenotype (Neri and Romana Di Raimo, 2010). But in some instances the concept of “type and countertype,” originally proposed by Lejeune (1966), may be invoked. Deletion of 7p15 may cause the cranial bones to fuse prematurely (craniosynostosis), due to abnormal behavior of osteoblasts at their periphery, whereas duplication leads to underdevelopment of the skull, with a large and confluent fontanelle (Stankiewicz et al., 2001c). Deletion of 15q26.1-qter (which removes the growth factor locus, IGFR1) is associated with intrauterine growth retardation, whereas dup(15)(q26.1-qter) may cause a syndrome of postnatal overgrowth (Nagai et al., 2002; Faivre et al., 2002).
Quantitative Assessment How do we determine what is a large or a small degree of genetic imbalance? First, we can take a quantitative approach—how much material is involved. The finer detail, as measured in megabases of DNA, is known for each chromosome band (Table 3–1). Now, in the second decade of this century, as microarray methodology becomes a firstline chromosomal investigation (Miller et al., 2010), the counselor will become familiar with deletion/ duplication reports couched in precise terms of start and finish basepair numbers. With respect to “classical” degrees of imbalance (several megabases), for practical purposes the blunter measurement of haploid autosomal length (HAL) is useful (see Appendix B). The largest chromosome, no. 1, comprises 8.4% of the HAL, whereas chromosome 21, the smallest, is 1.9%. As a very general rule, if the imbalance consists of less than 1% of HAL, the conceptus is often viable in utero, and live birth frequently results. If
Table 3–1. Actual Lengths of DNA Accommodated in Each Band, Measured in Bases, Using Chromosome 19 as a (Helpfully Small) Example BAND
BASES FROM PTER TO LOWER MARGIN OF THIS BAND
p13.3 p13.2 p13.13 p13.12 p13.11 p12 p11 cen q11 q12 q13.11 q13.12 q13.13 q13.2 q13.31 q13.32 q13.33 q13.41 q13.42 q13.43
8,362,000 bp 12,656,000 14,916,000 17,628,000 22,600,000 27,100,000 30,000,000
8,362,000 bp 4,294,000 2,260,000 2,712,000 4,972,000 4,500,000 2,900,000
32,500,000 36,900,000 40,211,000 43,221,000 46,833,000 49,541,000 51,046,000 54,357,000 58,872,000 60,979,000 63,688,000 67,000,000
2,500,000 4,400,000 3,311,000 3,010,000 3,612,000 2,709,000 1,505,000 3,311,000 4,515,000 2,107,000 2,709,000 3,312,000
SIZE OF THIS BAND
Notes: Cytogenetic resolution is at very high level (850-band). The p terminus is in band p13.3, and the q terminus in band q13.43. The whole chromosome is 67 Mb long, 30 Mb in the short arm and 47 Mb in the long arm. The average band size is approximately 3.5 Mb. Data from Internet site of the Weizmann Institute of Science.
the excess is greater than 2%, in utero lethality, with spontaneous abortion, is likely. Imbalance involving autosomal deficiency (partial monosomy) is generally much less survivable than is duplication (partial trisomy).
Qualitative Assessment Quantitative assessment of the amount of chromatin deleted or duplicated, based on classical cytogenetics, is rather crude. In general, dark G-bands are low in gene content, and light G-bands are gene rich; the telomeric regions have the highest gene density. More precisely, Saccone et al. (1999) have
The Origins and Consequences of Chromosome Pathology •
47
determined, on the basis of probing for regions rich in GC sequences, those parts of the different chromosomes that carry a greater quantum of genes (Fig. 3–10). Chromosomes, or chromosomal segments, of greater or lesser gene density are likely to have, in the unbalanced state, a lesser or greater survivability, respectively. Assessment is made, however, not on this theory, but upon the empiric observations of phenotypes. Some segments (e.g., 9p, all of 21) appear to have a substantial pre- and postnatal survivability in the trisomic state, whereas a lesser number of segments (e.g., distal 4p) are sometimes viable when monosomic. Chromosome 13 provides the most impressive examples of viability for a large autosomal imbalance. Trisomy for the whole of chromosome 13—fully 3.7% of the HAL —frequently goes through to live birth, and in the 13q–deletion syndrome, monosomy occurs for up to 2.5% of HAL. This presumably reflects a low gene density on this chromosome (Fig. 3–10), and a relative paucity of genes that are sensitive to dosage imbalance. The same likely applies to chromosomes 18 and 21. Occasionally, imbalance is so “small” that the effect on the child’s physical phenotype is only very minor, and intellectual function can remain within the normal range, albeit toward the lower end of that range. There are some segments which, when duplicated or deleted, appear to cause no abnormality at all. For example, trisomy for the segment 9p12p21.3 (comprising as much as ~0.6% of HAL) was found in a physically and mentally normal man, who had “learned a technical profession” (Stumm et al., 2002). The concept of heritable “euchromatic deletions and duplications without phenotypic effect” is discussed in Chapter 16. Other segments impart a serious trisomic/ monosomic effect even for a tiny amount of chromatin. Deletion of the single band 17p13.3, for example, causes the severe phenotype of the Miller-Dieker syndrome. The least number of loci that can be removed in a deletion is one; in Rubinstein-Taybi syndrome, for example, the loss of a single copy of the gene CREB within 16p13.3 suffices to produce the phenotype. Now, in the microarray era, a qualitative assessment may include, for smaller size segments (measured in kilobases), an illustration to accompany the cytogenetic report from one of the genome browser Web sites that shows the genes within that segment. Sometimes, a particular gene may present itself as a plausible, or perhaps the known basis of the 48
•
BA SIC CONCEPTS
associated phenotype; rather often, however, the list of genes will include several whose role, if any, in determining or contributing to the observed clinical picture may be quite obscure. K A R YOTYPE- PH EN OTYPE CO R R EL AT I O N S F OR D EL ET I O N S A N D D UPL I CAT I O N S
Differing lengths of deleted or duplicated segments enable a dissection of the specific segmental contributions to components of an abnormal phenotype. These various observations of karyotype-phenotype correlations permit judgment concerning whether a particular duplication or deficiency may be of minor, major, or lethal effect. A “malformation map” can be produced from documenting the association of certain congenital defects with particular segmental aneusomies (Brewer et al., 1998, 1999; Fig. 3–11). One of the most commonly seen defects is a heart malformation. The complex twisting and folding of the cardiac tube is, apparently, particularly vulnerable to an incorrect dosage of certain genes. These genes may reside in those chromosomal regions associated with heart defects, and van Karnebeek and Hennekam (1999) have documented these associations. A “susceptible cardiological karyotype” does not, however, necessarily determine that a heart defect will happen, as attested, for example, by the observation of discordant monozygous twins with a dup(4q28.3-qter) (Celle et al., 2000). The brain is uniquely susceptible to chromosomal imbalance, and Tyshchenko et al. (2009) have constructed a map. We have undertaken similar phenotype mapping studies with respect to kidney defects (Amor et al., 2003), and to epilepsy (Singh et al., 2002a), hypothesizing that the chromosome regions thus illuminated may serve as candidate regions for the discovery of renal and epilepsy genes. The application of microarray analysis enables a finer focus to be taken. Catelani et al. (2009) have looked for “microimbalances” in children with syndromic deafness, anticipating that these might point the way to the discovery of hearing genes (Table 3–2). Kirchhoff et al. (2009) describe correlations in the distal half of chromosome 13 on the basis of microarray analysis, associating particular traits with certain “microsegments” (Fig. 19–2). Not that a one-to-one connection between a deleted/duplicated segment and a specific trait can necessarily be drawn; and for example, we have proposed that the particular nervous system malformation of periventricular
FIGURE 3–10 Gene-rich regions of the human karyotype. The bands in red hybridize to genomic DNA abundant in CpG islands. Note the absence or near-absence of gene-rich regions in the three “viable” autosomes, nos. 13, 18, and 21 (and cf. the reference on p. 278 to the mere 225 genes on chromosome 21). Chromosome 19, on the other hand, is well colored in, which accords with knowledge of the rarity of viable chromosome 19 imbalance. (From Figure 2 in S. Saccone et al., 1999, Identification of the gene-richest bands in human prometaphase chromosomes, Chromosome Research 7:379–386. Courtesy G. Bernardi; reproduced with the permission of Kluwer Academic Publishers.) See also separate color insert.
FIGURE 3–11 A duplication-malformation correlation map. Some chromosomal regions, in the duplicated state, are particularly associated with certain types of malformation. Presumably, these regions harbor genes that have roles in the formation of these particular organs. Other regions (including all of chromosome 19) are unrepresented, and some of these may contain “triplo-lethal genes.” ACC, agenesis of the corpus callosum; ASD, atrial septal defect; AVSD, atrioventricular septal defect; PDA, patent ductus arteriosus; VSD, ventricular septal defect. (From C Brewer et al., 1999, A chromosomal duplication map of malformations: regions of suspected haplo- and triplolethality—and tolerance of segmental aneuploidy—in humans, American Journal of Human Genetics 64:1702–1708. Courtesy D. R. FitzPatrick; reproduced with the permission of the University of Chicago Press.) A similar map has been drawn for deletions (Brewer et al., 1998).
Table 3–2. An Example of the Precision That Microarray Analysis Allows, and How the Data May Be Presented GENOMIC POSITION, Mb FROM PTER
del(1)(q23.3q25.2) dup(2)(q22.2q23.3) dup(6)(p25.2p25.3) del(11)(q13.2q13.4) dup(2)(q12.3q12.3) del(4)(q23q24) del(7)(q31.1q31.1) del(15)(q15.3q15.3)
PROXIMAL BREAKPOINT
DISTAL BREAKPOINT
160.19 . . . 161.21 143.67 . . . 144.59 2.98 . . . 2.86 67.55 . . . 68.07 107.03 . . . 108.13 101.15 . . . 101.51 109.93 . . . 110.06 41.41 . . . 41.68
175.27 . . . 175.56 150.78 . . . 150.86 1.26 . . . 1.32 70.31 . . . 70.60 108.34 . . . 108.50 101.96 . . . 103.22 110.31 . . . 110.39 41.85 . . . 42.76
MINIMUM SIZE (Mb)
14.06 6.19 1.60 2.24 0.21 0.46 0.25 0.17
ORIGIN
de novo de novo de novo de novo maternal paternal maternal maternal
Notes: Measurements in boldface indicate breakpoints for the minimum determined size of the deletion/duplication; nonboldface indicates maximum size. These “microimbalances” were identified in a group of children with syndromic hearing loss, assessed at the Genetic Counseling Service of the University of São Paulo, whose standard karyotypes had been interpreted as normal (Catelani et al., 2009). The deleted/ duplicated microsegments in the de novo cases presumably included genes whose imbalance contributed to the other phenotypic traits manifested by these children, but which might also have included a “hearing gene,” for which a dosage imbalance could have compromised function of the gene product. For example, the deletion of chromosome 1 included DFNA7, a gene which is the basis of one form of dominantly inherited nonsyndromic deafness, and DFNA7 haplo-insufficiency might have contributed to the child’s profound deafness. The case in those having inherited the imbalance from a normal parent is less secure; these could be normal copy number variants.
nodular heterotopia might be an epiphenomenon accompanying a number of microdeletion syndromes, rather than the direct consequence of specific segmental imbalances (van Kogelenberg et al., 2010). For the most part, the clinical states due to chromosomal imbalance are fixed and static. Structural defects such as a cardiac septal defect, or facial dysmorphism, are not progressive (although they may be evolving) conditions: they were established during embryogenesis and fetal development, and, in essence, will stay that way. They may, of course, set the stage for consequential progressive change, such as a urinary tract defect that has back-pressure effects upon a kidney, affecting renal function; but this is a secondary factor. The brain, the most vulnerable organ, is similarly fixed in terms of its underlying anatomy, and chromosome disorders would not typically be described as neurodegenerative. The most notable exception to that rule is the long-recognized dementia that typically commences around age 40 in Down syndrome, and which reflects the effects of a triple dose of the amyloid precursor protein gene on chromosome 21, with a gradual accumulation in the brain of the abnormal protein. Other exceptions exist. De Bruijn et al. (2010) describe a de novo apparently balanced
translocation in which no genes were lost, but the new position in the genome of a “brain gene” (LRFNS) led to its much diminished activity (thus, an epigenetic effect), and this was associated with an actual loss of acquired skills in mid-childhood. The cognitive deterioration in the ring 20 syndrome may reasonably be described as neurodegenerative, albeit that the onset of epilepsy may be a necessary aggravating factor. Database Resources Two important hard-copy sources provide information on, first, the clinical features of specific duplications and deficiencies, and second, viability of a particular segment. Schinzel has compiled his Catalogue of Unbalanced Chromosome Aberrations (2001), an invaluable resource documenting the clinical phenotype in about 2000 different aneuploid states that are compatible with live birth, and many of which are associated with survival through to early childhood or beyond. Stengel-Rutkowski et al. (1988) have gathered data from 1120 translocation pedigrees, determining, for numerous segments in the partially trisomic and partially monosomic states, the likelihood that a pregnancy
The Origins and Consequences of Chromosome Pathology •
51
would proceed through to live birth (referred to in detail in Chapter 5). It is an obvious point, but worth restating: the defect in these aneuploid states involves too much or too little of what is normal chromosome material. The “third” chromosome in standard trisomy 21 is a perfectly normal chromosome 21, with a perfectly normal complement of chromosome 21 genes. How, therefore, could it be that an additional amount of normal genetic message leads to an abnormal interpretation of that message? This is one of the great remaining unanswered questions of biology, which we touch upon (no more than that) in Chapter 18. THE SEX C H RO MOSOM ES
Sex chromosome imbalances need to be considered separately. Any X chromosomes in excess of one are genetically inactivated. Thus, indicating the inactivated X in lowercase, normal females are 46,Xx, normal males are 46,XY, Turner females are 45,X, Klinefelter males are 47,XxY, and other X aneuploidies are 47,Xxx, 48,Xxxx, 48,XxYY, 49,XxxxY, 49,Xxxxx. As for the Y chromosome, its active genetic material is confined to only a small segment, these genes being mostly related to sex determination and testicular function. Thus, in spite of the presence of one or more whole X or Y chromosomes in excess in the 47-, 48- and 49-chromosome states, in utero survival remains possible. Indeed, for 47,XXX, 47,XXY, and 47,XYY, survival is apparently quite uncompromised. Gonadal development in X aneuploid males is particularly affected, and intellectual function is jeopardized to a mild or moderate extent in the n ≥47 states in both sexes. 45,X has a high in utero lethality, although the small fraction surviving to term as females with Turner syndrome show, in contrast, a remarkably mild phenotype.
other chromosomal imbalances, are described (see Chapter 22).
THE MOSAIC STATE Whether mosaicism matters depends upon which tissue, and how much of that tissue, is abnormal. If a majority of the soma is chromosomally abnormal, the phenotype is likely to be abnormal. If only a tiny fraction of some tissue were involved, in which the aneuploidy would have essentially no effect—if, say, some of the bony tissue of the distal phalanx of the left little toe were trisomic 21, and the rest of the person 46,N—it would never be known. Indeed, as mentioned earlier, it could be everyone has mosaicism, for the most part harmless, in certain tissues or organs. However, in regard to disease, a very minor degree of mosaicism could still be important if a crucial tissue carried the imbalance. An abnormal chromosome confined to tissues of, say, a localized area or cell type in one part of the brain could theoretically cause neurological dysfunction. (Perhaps twenty-first century technology will devise a “functional cytogenetic MRI scan” that could map out brain regions with an aneuploid chromosomal complement!) Abnormality involving a gonad or part of a gonad (“gonadal mosaicism”) could lead to a child being conceived with that aneuploidy. Mosaicism confined to extraembryonic tissue may be without phenotypic effect, although it certainly causes anxiety if it produces an abnormal test result at prenatal diagnosis. Confined placental mosaicism (CPM) may exist unbeknown in pregnancies producing normal infants, as Lestou et al. (2000) showed in a study of 100 placentas, with five revealing CPM for trisomies 2, 4, 12, 13, and 18. Mosaicism may frequently be observed at the IVF laboratory in the early cleavage embryo, and of spectacular degree, with different cells having different aneuploidies; a state of affairs that becomes very relevant in preimplantation genetic diagnosis (Chapter 26).
P HE NO CO P IES
Similar phenotypes may flow from different genotypes. “Pseudotrisomy 13” may be an autosomal recessive condition (Amor and Woods, 2000). The expression “DiGeorge syndrome” refers to an ensemble of signs that characterize the 22q11 deletion. Somewhat similar clinical pictures can be seen in deletions of 10p13 and of 4q34.2. Syndromes resembling Silver-Russell syndrome, Prader-Willi syndrome, and Angelman syndrome, but due to 52
•
BA SIC CONCEPTS
Mosaicism for a Full Aneuploidy As a general principle, an individual with an aneuploid line in only some tissues is likely to have a less severe but qualitatively similar phenotype to someone with the nonmosaic aneuploidy. The ascertainment of these individuals is biased: those with a more obvious phenotypic defect are, naturally, more likely to be detected. Mosaic Down syndrome— 46,N/47,+21—can be less obvious than standard
trisomy 21, and with a lesser compromise of intellectual function. The existence of 46,N cells in some of the brain tissue presumably has a moderating effect. Some aneuploidies can only, or almost only, exist in the mosaic state, the nonmosaic form being lethal in utero. Examples of this are 46,N/46,+8 and 46,N/47,+9 mosaicism. If the distribution of the aneuploid cell line is asymmetric, body shape may be asymmetric, generally with the hypoplasia in regions of aneuploidy. De Ravel et al. (2001) described hemifacial microsomia (one side of the face being underdeveloped) and other body asymmetry in two children with autosomal mosaicism, one for trisomy 9 and the other trisomy 22. The child with 47,XY,+22/46,XY had 9/10 cells +22 on skin fibroblasts from the arm on the right (underdeveloped) side, compared with 5/11 on the left arm (blood karyotype was 46,XY). Molecular analysis supported there having been a postzygotic anaphase lag that had produced the 46,XY line from an initially 47,XY,+22 conception. A surprising case is that of Greally et al. (1996): a child with mosaic trisomy 16, a cardiac malformation, and otherwise (barring a unilateral simian crease) not dysmorphic, and her neurodevelopmental progress has been quite normal. One might suppose (but could not prove) that the trisomic cell line was somewhat confined in distribution and excluded the brain. Mosaicism excluding the bone marrow will give a normal blood karyotype, and vice versa mosaicism confined to marrow would be seen on routine peripheral blood analysis, but not on other samplings; mosaic trisomy 8 may provide examples in both directions. Examples of presumed very lowlevel trisomy mosaicism have come to light through prenatal diagnosis, such as trisomy 13 mosaicism in an apparently normal child with 1 cell out of 400 on cord blood (Delatycki et al., 1998). In sex chromosome mosaicism, fertility can exist when otherwise infertility is the rule: for example, in “formes frustes” of Turner syndrome with 45,X/46,XX and of Klinefelter syndrome with 46,XY/47,XXY.
Mosaicism for a Structural Rearrangement We reviewed a three-decade experience in New Zealand, and only 12 cases of mosaicism for a structural rearrangement had ever been recognized, of which at least 3 were regarded as balanced, and 8 presumably unbalanced. This equated, crudely, to an incidence of around one case per year per 1,000,000
population (Gardner et al., 1994). This is likely to be a considerable underestimate. Leegte et al. (1998) reviewed the literature and could record 29 cases of mosaicism for a balanced reciprocal translocation. This state, presumably, would be without any phenotypic effect. The only practical implication would be if the mosaicism extended into the gonad, an example of which is provided by Shapira et al. (1997c). A man, himself quite normal, was mosaic for a pericentric inversion, 46,XY/46,XY,inv(9) (p24q34.1), and he had a child with a recombinant chromosome that had caused a deletion 9p/duplication 9q syndrome. Some cases of “balanced” translocation mosaicism will have been ascertained through the phenotypic abnormality of a proband, and it is arguable whether the rearrangement had caused the abnormality or was coincidental (Aughton et al., 1993). With an unbalanced karyotype, the broad rule applies that the mosaic form is likely to be less severe than the nonmosaic. Pigmentary skin anomaly is a notable and clinically useful phenotypic trait that can characterize this type of unbalanced mosaicism, the important categories being hypomelanosis of Ito, linear and whorled nevoid hypermelanosis, and “phylloid” (leaf-like) pigmentary disturbance (Verghese et al., 1999; Riberio Noce et al., 2001). The distribution of the abnormal cells in hypomelanosis of Ito, and thus of dyspigmentation, follows the lines of Blaschko, and Magenis et al. (1999) use the expression “Blaschkolinear malformation complex.” Niessen et al. (2005) studied in some detail a girl with three shades of skin pigmentation— hypopigmented, normally pigmented, and hyperpigmented (“cutis tricolor”)—following the lines of Blaschko. She karyotyped 45,X on blood, and 47,XX,+7 on skin biopsied from the darker skin. Asymmetry is a further clinical pointer (Woods et al., 1994). “Functional mosaicism” for a structural rearrangement is exemplified by the X-autosome translocation in which different regions of the body have differing ratios of inactivation of the translocation and the normal X, and this, also, can lead to hypomelanosis of Ito (Hatchwell et al., 1996). An interesting category of mosaicism for a structural rearrangement is that in which two lines of opposite imbalance coexist, with or without a normal cell line as well. Here, the error must have happened at a very early stage, and quite possibly, in those cases lacking a normal cell line, at the very first mitosis of the zygote. Such a case is described in Morales et al. (2007a), who analyzed a boy with the
The Origins and Consequences of Chromosome Pathology •
53
karyotype at birth of 46,XY,del(7q)/46,XY,dup(7q), although by age 12–14 months, the deletion cell line had disappeared, at least from blood and exfoliated urinary epithelial cells. Presumably, the karyotype at conception was 46,XY; but then the two chromosome 7 homologs underwent an unequal exchange of q21.1-31.3 material, generating, in the two daughter cells from the first mitosis, the deletion and duplication lineages. If, say, the error occurred one division later, at one of the two second mitotic divisions, a normal cell line might be retained, according to which progeny cells then came to comprise the inner cell mass. Tissue Sampling in the Detection of Mosaicism. As already noted, mosaicism can, in theory, be very widespread, and the distribution of the different cell lines can vary considerably. Analysis of tissues other than blood can clarify the picture: readily accessible tissues such as buccal mucosal cells and urinary epithelial cells, and infrequently available material from posttermination or postmortem studies. Kingston et al. (1993) described a fetal study in which several tissues taken posttermination had various fractions of mosaicism for an additional abnormal chromosome, including 88% of brain cells, while only 3% of amniotic fluid cells and no cells from a sample of fetal blood had the abnormal chromosome. Reddy et al. (1999) studied a retarded woman with mosaicism for an “add(3),” whose blood karyotype proved to be 46,XX,der(3)t(3;14)(q29;q31)/46,XX. Using a 14q subtelomeric probe, they could show that 86% of buccal cells showed three FISH signals, and so contained the der(3), while the 14% of cells with two signals were normal. (In fact, this ratio was very similar to that of the peripheral blood, which was 83:17.)
Gonadal (and Somatic-Gonadal) Mosaicism The classical view is that gametes with a chromosomal abnormality are typically produced by 46,N parents, whose gonads are chromosomally normal. The abnormality is presumed to have arisen at meiosis and to have affected only the gamete(s) arising from that single meiosis. If, however, an abnormality had arisen during formation of a germ cell prior to the onset of meiosis, an abnormal cell line can become established and occupy a part of the gonad or gonads. This is gonadal mosaicism.
54
•
BA SIC CONCEPTS
Gonadal mosaicism is suspected upon the observation of a chromosomally normal couple (on blood testing) having had two children with the same abnormal karyotype. Molecular analysis can allow an inference of who is the carrier parent, such as Tosca et al. (2010) show in the family study of two children with a dup(4)(q22.2q32.3), in which the microsatellite pattern indicated a maternal origin. Definitive proof is provided by analysis of gametes. For example, in a case that had come to notice through an IVF clinic, Somprasit et al. (2004) report a couple having had a 21q duplication in two embryos subjected to preimplantation genetic diagnosis (PGD), and then, using FISH, showed the same duplication in 6.6% of 1002 of the father’s sperm. The abnormality was not present in his blood. The direct testing of ovarian tissue is noted below. Cells destined to give rise to gametocytes originate from the yolk sac in early embryogenesis and migrate to the gonadal ridge on the dorsal wall of the abdominal cavity, where, along with the supporting cells, they come to comprise the tissue of the gonad. In doing so, gametocytes must replicate many times, going through about 30 cycles of division. Thirty cycles produces 230 (about 1,000,000,000) progeny cells, and the potential for error exists at each cell division contributing to this population. These errors could be nondisjunctions or the production of structural rearrangements. Edwards (1989) offers a startling insight into the actuality of gonadal mosaicism. He points out that, in the male, the total length of seminiferous tubule is about 1 kilometer. If a mutation were to occur in a spermatogonium in, say, the twentieth cycle of division, its progeny would then go through 10 more cycles and comprise 210 (about 1,000) cells. This would be only a millionth (1000/1,000,000,000) of the 1 km of tubule—a mere 1 mm. So a man mosaic in such a way would have a risk of only 1 in a million to father a conception with this particular abnormality. From similar reasoning, a defect arising at the tenth cycle could affect 1 m of tubule, and thus carry a risk of 1 in 1000. Oögonia need go through a lesser number of cycles, but the same principles broadly apply. If the abnormality arose in embryogenesis prior to the differentiation of the germ cell line, the soma may also be involved: this is somatic-gonadal mosaicism. Sachs et al. (1990) demonstrated ovarian mosaicism by direct gonadal samplings in a woman who had had one Down syndrome child and three
other trisomic 21 pregnancies; she had already been shown as a (very low-level) somatic mosaic, her blood karyotype 46,N[97%]/47,+21[3%]. Tissue cultured from ovarian biopsies showed almost half the cells in each ovary to be 47,XX,+21. Other such cases are mentioned on p. 282. Hultén et al. (2008), from an analysis of fetal ovaries, suggest that ovarian mosaicism may be a great deal more frequent than has been recognized, and they make the challenging, indeed startling statement that “parental aneuploidy gonadal mosaicism may be the major underlying reason for trisomy 21 conceptions” (and see earlier discussion). Gonadal mosaicism can also arise due to “correction” of an initially trisomic conception (Fig. 3–8). The classic example is mosaic trisomy 21, in which a 47,+21 embryo discards the extra chromosome in a cell which goes on to produce most tissue lineages, but not in the cell destined to give rise to gonadal tissue (Kovaleva, 2010). The female embryo may engage this process more efficiently than the male. The clinical observation supporting the interpretation is that the mosaic mothers (who presented due to having had a nonmosaic DS child) are of a typical
maternal age range, whereas their mothers—the grandmothers of the DS children—were of older maternal age at the time their daughters had been born. Figure 3–12 shows an example of somaticgonadal mosaicism for a structural rearrangement. The index case was identified with a small intrachromosomal del(1) at routine prenatal diagnosis. The father was mosaic for this deletion, in 20% of lymphocytes. Of his two other children, one had normal chromosomes, and the other had the same deletion. The father is phenotypically normal, and the older child with the deletion has an IQ in the low normal range. A similar circumstance is recorded in Fan et al. (2001): a university-educated man working as a financial planner, having the blood karyotype 46, XY[24]/46, XY,dup(8)(p21.3p23.1)[6], fathered two children with 46,XX,dup(8) (p21.3p23.1). These girls had poor language development, clumsy motor abilities, and minor facial dysmorphism. If the proportion of abnormal cells in the mosaic parent was higher or differently distributed, that parent may manifest some signs of the partial aneuploid state. The father reported in
(a) N
(b)
1
1
1
Normal cell line
1
1 del(1)
1 del(1)
Abnormal cell line
FIGURE 3–12 A family exemplifying somatic-gonadal mosaicism. (a) Pedigree. The father had the mosaic karyotype 46,XY,del(1)(q25q31.2)[16]/46,XY[4] on lymphocyte study. Two children have the del(1) (q25q31.2) in nonmosaic state. The family was ascertained following routine prenatal diagnosis. The older sibling’s development was judged, at age 5 years, to be in the low average range; height, weight, and head circumference were in the range 20–25th centiles. The father worked as an electrician. (b) Partial karyotype showing the father’s two cell lines: two normal no. 1 chromosomes, and one normal and one deleted chromosome 1. The segment 1q25-q31.2 is shown crosshatched. (Case of G. Dawson.)
The Origins and Consequences of Chromosome Pathology •
55
Kennedy et al. (2001) had a dup(8)(p23.1p23.1) in mosaic state, in the ratio normal:duplication of 17:8 on blood sampling, and he himself had a heart defect, as did his nonmosaic dup(8) daughter. Her defect was, however, rather more severe than her father’s: she had a fairly complex malformation, including a right-sided aortic arch, while he had only a right-sided arch. Notably, the daughter was described as “achieving top grades in school,” a very unusual phenotypic commentary in a child with a nonmosaic chromosome duplication.
Placental and Amniotic Fluid Mosaicism About 1%–2% of placentas can have a different chromosomal constitution from that of the embryo, with usually the embryo being normal and the placenta trisomic. This is “confined placental mosaicism.” Thus, in 1%–2% of chorionic villus sampling (placental biopsy) there will be a potentially misleading result. Fortunately, these uncommon instances can, as a rule, be recognized as such, although not without causing some anxiety at the time. In a few confined placental aneuploidies, the function of the placenta may be compromised, and fetal well-being may be affected. Infrequently, true mosaicism is recognized at amniocentesis. Occasional cells with a chromosomal abnormality, if they are solitary or involving a single clone, are generally regarded as having arisen in vitro (“artifactual mosaicism”). At least most of the time, this is probably the correct interpretation. We consider placental and amniotic fluid cell mosaicism in detail in Chapter 27.
QUALITATIVE IMBALANCE The idea that abnormality could be due to unequal parental contributions of an overall correct amount of chromosome material seemed most remarkable in 1980 when Engel first made the suggestion and coined the expression “uniparental disomy.” It has come to be accepted fact. The two disorders that, par excellence, exemplify the concept of qualitative imbalance are Prader-Willi syndrome (PWS) and Angelman syndrome (AS). The concept of genomic imprinting, discussed earlier, is central to an understanding of the etiology. Each syndrome is due to the nonexpression of different (but neighboring) segments within the proximal long arm of chromosome 15. A “PWS critical region” is normally 56
•
BA SIC CONCEPTS
expressed from only one chromosome, in this case the paternally originating chromosome. The maternal-originating region is normally inactive and alleles in this region are not transcribed. Thus, there is a “functional monoallelism.” If the paternal PWS region is absent, the maternal one cannot “fill the gap,” and the consequential functional nullisomy is the root cause of PWS. An “AS critical region” exists, lying just a little distal from the PW region. Likewise, it needs only monoallelic expression for normal phenotypic function. In this case, the maternal region is active, and the paternal region, having been imprinted, is inactive. If the maternal region is absent, there can be no genetic activity, and this causes the AS phenotype. Absence of the paternal PWS region or maternal AS region can flow from two major mechanisms. First, in uniparental disomy (UPD), one parent fails to contribute a chromosome 15; and the “correcting” presence of two copies from the other parent cannot restore a proper balance. This can be heterodisomy (the two homologs being different), or isodisomy (they are identical). Second, there can be a deletion within proximal 15q that removes a segment of chromatin containing the PWS and AS regions. These issues are dealt with in some detail in Chapter 22. As the imprinting story has evolved, it has emerged that several, indeed most chromosomes, appear not to be subject to imprinting. For these chromosomes, and with both homologs equally genetically active, regardless of the parent of origin, UPD will have no untoward effect. Only if the UPDcontributing parent should happen to be heterozygous for a recessive gene, and if this is the isodisomy category of UPD, will the child be affected, displaying the condition concerned, due to homozygosity (“isohomozygosity”) for that recessive gene. Rare instances of this scenario are known. Similar considerations may apply in the trisomies. Naturally, one parent must have contributed more than one homolog. Considering the example of Down syndrome, does the parent from whom the disomic gamete came contribute two different chromosome 21s? In other words, does the child inherit a chromosome 21 from three of the grandparents— “heterotrisomy”? Or does the parent contribute two identical (isodisomic) chromosome 21s? Whether phenotypic differences may flow from these different possibilities is uncertain, although Baptista et al. (2000) suggest that heterotrisomy 21 may, of itself, convey a greater risk for a specific heart
malformation, ventricular septal defect, speculatively due to a damaging interaction of three subtly different protein products from a 21q “heart locus.” Uniparental disomy for the entire chromosome set—“uniparental diploidy”—has a devastating effect on development. If a conceptus has lost its maternal complement, and the paternal complement is doubled, embryonic development arrests, leaving only grossly abnormal chorionic villi comprising the pregnancy. This is a hydatidiform mole (p. 394). If a 46,XX ovum at meiosis I attempts a parthenogenetic development, a grossly disorganized mass of embryonic tissue results: an ovarian teratoma. If a triple set of chromosomes (triploidy) is present at conception, there is either a diploid maternal set plus a haploid paternal set, or vice versa. These different parental origins determine very different abnormal fetal and placental phenotypes (p. 287). SEG M EN TAL UN IPA RE NTA L DI SOMY
A mitotic mechanism that can lead to functional imbalance, if the segments exchanged are in a region subject to imprinting, is somatic recombination. The first shown example of this causing a dysmorphic syndrome is the segmental paternal uniparental disomy for 11p that underlies some BeckwithWiedemann syndrome, 11p being a segment that is normally maternally imprinted. In the partial UPD(pat) cell line, this segment will now be expressing biallelically at distal 11p, instead of the normal monoallelic expression. The asymmetry of body growth in this syndrome reflects the distribution of two cell lineages: the normal biparental disomic line, and the functionally imbalanced UPD(pat) line.
SPORADIC AND RECURRENT ABNORMALITIES Chromosomally normal parents can produce abnormal gametes by nondisjunction, or by one of the other mutational mechanisms we have discussed earlier. The combination of factors that causes these defects in an individual case is unknown. No convincing case has ever been made for the agency of diet, illness, chemical exposure, or “lifestyle factors” in maternal chromosome 21 meiotic nondisjunction (Chapter 28), nor is there much support from epidemiological studies (Chapter 24). Noting the similarity of Down syndrome prevalence rates
worldwide, Carothers et al. (1999) comment that “the totality of published data could well be consistent with no real variation at all, and [this] might explain why a search for environmental factors associated with Down syndrome has been so unproductive.” The maternal age effect is of course important, indeed central, and any search for causes of chromosomal aneuploidy must take it into account. A plausible view is that there is a natural degeneration of the oöcyte, as we discussed earlier, and with reference to Figure 3–7. Simply put, eggs get older, and they show their age. (But we do not ignore the view of Hultén et al. (2008) that maternal gonadal mosaicism, with delayed maturation of the trisomic 21 oöcytes, is an alternative explanation.) Chromosomes are plastic, dynamic entities, and cell division is a complex mechanical process; and these qualities alone may suffice to endow the vulnerability that causes human aneuploidy and rearrangement. Given the assumption that all persons with intact gametogenesis are capable of producing an abnormal gamete, one view is that it may simply be that a certain background abnormality rate is intrinsic to the human species, and it is a chance matter whether this or that couple will have the misfortune to conceive the abnormality which, inevitably, someone has to bear. PARENTAL PREDISPOSITION TO N OND I S JUN CT I O N ?
An alternative view is that some 46,XX and 46,XY people are more prone than others to produce chromosomally unbalanced gametes. An intrinsic fault, or at least a vulnerability, in the mechanism of chromosome distribution at cell division could be the basis of the rare examples of recurring defects. The synaptonemal complex gene SYPC3 has been mentioned earlier; another candidate is the mismatch repair (MMR) gene, with particular reference to MLH1 (otherwise familiar to the counselor in hereditary nonpolyposis colon cancer) and MLH3, and the related meiosis genes MSH4 and MSH5 (Baarends et al., 2001; Lenzi et al., 2005; Terribas et al., 2010). Given the complexity of the apparatus and process of cell division, it is logical that errorcausing mutants in the controlling genes (regardless of whether this might include any of the aforementioned) would exist. Whether there might be milder alleles at postulated cell-division loci, which could more widely be the cause of occasional nondisjunction, remains a matter for speculation.
The Origins and Consequences of Chromosome Pathology •
57
FIGURE 3–13 Chromosomes at synapsis exist as double-chromatid structures (e.g., the reciprocal translocation quadrivalent at right). But, for simplicity, we generally represent them with just the one chromatid (left).
Nevertheless, a geneticist could scarcely ignore that there might exist subtle genetic variation potentially setting the stage for nondisjunction. A Note on the Diagrams. Following the progress of rearranged chromosomes during meiosis is
58
•
BA SIC CONCEPTS
not easy, so we have taken some liberties in simplifying the diagrams. Most of these diagrams depict the synapsing chromosomes at meiosis with just one chromatid; of course, the chromosome has actually replicated at this point and exists as a double chromatid entity (Fig. 3–13).
4 deriving and using a risk figure
RISK IS A CENTRAL CONCEPT in genetic counseling. By risk, we mean the probability that a particular event will happen. Probability is conventionally measured with a number ranging from 0 to 1. A probability (p) of zero means never, and a probability of 1 means always. For two or more mutually exclusive possible outcomes, the individual probabilities sum to 1.0 (or 100%). Thus, someone who is a heterozygote for a particular rearrangement might, in any given pregnancy, have a probability of 0.10 (10%) of having an abnormal child and a probability of 0.90 (90%) of having a normal child. We may speak in terms of risks of recurrence or of occurrence: the probability that an event will happen again, or that it will happen for the first time. Risk can also be presented as odds: the ratio of two mutually exclusive probabilities. The odds for the preceding hypothetical heterozygote would be 9:1 in favor of a normal child. The work risk has two important meanings in the English language. First, there is the scientific sense of probability that we already discussed. Second, as most people use the word, it conveys a sense of
exposure to danger. Our hypothetical heterozygote runs the risk that an unfortunate outcome may occur (an abnormal child, or an abnormal result at prenatal diagnosis). In the genetic counseling clinic these meanings of risk coalesce in some ways, to which the counselor needs to be sensitive. We might instead use such everyday words as chance or likelihood, which have no negative connotation, to refer to the fortunate outcome of normality. The words fortunate and unfortunate are also chosen deliberately: the wanted or the unwanted event will occur entirely by chance, analogous to tossing a coin, throwing a dice, or being dealt a card.
Different Types of Risk Figure Geneticists arrive at risk figures in a number of ways (Harper, 2010), two of which have particular application to cytogenetics. 1. Empiric risks. In the great majority of chromosomal situations, no clear theory exists from which •
59
the risk can be derived, and one must observe what has happened previously in (as far as one can judge) the same situation in other families and make an extrapolation to the family in question. Empiric risks thus appeal to experience, and they only estimate the intrinsic, true probability. The data may be available in the literature record or in specific databases; or the counselor may need to derive a “private estimate” from an analysis of the client’s family. The risk estimate has a greater or lesser degree of precision, depending on how much data have been accumulated upon which the estimate is based. 2. Mendelian risks. If a clear model of inheritance is known, risk figures derived by reference to that theory may be used. In practice, only Mendel’s law of segregation is applied in this context. When a pair of homologous chromosomes segregates at meiosis, it is a random matter which chromosome enters the gamete that will produce the conceptus. Each has an equal chance: a probability of 0.5. As an example, the X chromosomes in the fragile X heterozygote, the normal X and the fragile X, display 1:1 segregation. In the microarray era, the 1:1 segregation of copy number variants becomes of relevance. This is assumed to be a true risk, not an estimate: it is 0.5 exactly. Consider, for example, the common situation of a young couple having had a child with Down syndrome. Nothing is known about nondisjunction that could provide a theoretical model on which to base a recurrence risk figure. We therefore use empiric data, that is, information obtained from surveying large numbers of other such families. It may be observed, for example, that in these families about 1 pregnancy in 100, subsequent to the index case of Down syndrome, produced another child with Down syndrome. Formally expressed, this is a segregation analysis. From this rate of 1/100 we can derive a risk figure of 1%, which we then have as the basis for advising patients. (Actually, it is not quite as straightforward as this in Down syndrome; see Chapter 18.) Likewise for the circumstance of the parent heterozygous for a chromosomal rearrangement, the counselor can consult data that have been accumulated by workers in the field, foremost among whom, in respect of reciprocal translocations, are Stengel-Rutkowski et al. (1988) and Cohen et al. (1992, 1994), and more recently, Midro et al. (2000). Since almost all reciprocal translocations are unique to one family, it is not necessarily simple to estimate a figure for a family with a “new” 60
•
BA SIC CONCEPTS
translocation, but an attempt can be made (see Chapter 5). On the other hand, for the Robertsonian translocations, each type of which can generally be regarded as the same between families, extrapolation of risk figures from historical data to a current family is usually valid. Hook and Cross (1982) note the importance of distinguishing between the rate (which may be thought of as “past tense”) and the risk (which is “future tense”). They emphasize that, while geneticists routinely extrapolate from rates in one population at one point in time, and may use these figures as risk estimates in another population and certainly at a later point in time, they should be on their guard for any evidence that a condition varies with time, geography, or ethnicity. But actually, there is little indication that any important variation exists: chromosomal biology appears to be rather consistent throughout the human race (p. 406).
Doing a Segregation Analysis Segregation analysis is essentially a simple exercise. A farmer who surveys a flock of newborn lambs and notes that 3 are black and 97 are white has done a segregation analysis. In human cytogenetic segregation analysis, the exercise involves looking at a (preferably large) number of offspring of a particular category of parent: parents who carry some particular chromosome rearrangement or those who have had a child with a chromosomal abnormality, they themselves being karyotypically normal. The proportion of these parents’ children who are abnormal is noted (say, 3 out of 100), and this datum serves as the point estimate of the recurrence risk (thus, 3%). Although segregation analysis is simple in principle, there are potential pitfalls in its application, the most important of which is ascertainment bias. We will deal with this problem only briefly. It is important that the counselor know of ascertainment bias and recognize whether it has been accounted for in the published works consulted. But it is not necessary to understand the complex and sophisticated mechanics of segregation analysis in detail. The reader wishing fuller instruction is referred to Murphy and Chase (1975), Emery (1986), and Stene and Stengel-Rutkowski (1988). The classic example of ascertainment bias is that of the analysis of the sex ratio in sibships of military recruits in World War I. Adding up the numbers of brothers and sisters, there was a marked excess of males. But of course (in 1914–1918) the recruit himself had to
be male. Once he was excluded from the total in each sibship, the overall sex ratio was normal, namely, 1.0. Likewise, in a cytogenetic segregation analysis, the individual whose abnormality brought the family to attention—the proband—is excluded from the calculation. That person had to be abnormal. Furthermore, that individual’s carrier parent, grandparent, and so on in a direct vertical line had to be (almost always) phenotypically normal to have been a parent. These individuals must also be excluded from an analysis of their own sibship, if that generation is available for study. Other sibships may be included in full. These manipulations—dropping the proband and the heterozygous direct-line antecedents—are the major steps to be taken to avoid the distorting effects of ascertainment bias. Another potential methodological confounder for the aficionado is ascertainment probability. For example, families with more affected members may be more likely to come to medical attention, which would unduly weight the data. There are means to overcome this problem. Essential to a good analysis is good data, or at least as good as possible. Some retrospective information may be uncertain. In a family translocation study, did a phenotypically abnormal great uncle who died as a child in 1930 have the “family aneuploidy”? (Old photos may be very helpful in this respect.) Some family skeletons may remain in cupboards unopened to the interviewer. Particularly in the follow-up of prenatal diagnosis results, it is important to know the endpoint of data collection of the child and how the data were collected: at birth, or until school age; by formal examination, or by anecdotal report. The investigative zeal, clinical judgment, and personal qualities of the researcher are crucial in getting the right information, and getting it all.
phenotypically abnormal offspring. The figures in parentheses give raw totals in these sibships, but then the proband (IV:4) and his heterozygous antecedents (II:1 and III:1) are excluded from their sibships. Thus, we have the following:
PHENOTYPICALLY NORMAL PARENT OF AFFECTED CARRIER NON- UNKARYOSIBSHIP CARRIER TYPED
I:1 II:1 II:2 III:1 III:2 III:7 Total
0 1 0 2 (3) 0 0 3
1 (2) 1 (2) 1 0 1 0 4
2 0 0 1 0 1 4
0 2 1 0 0 0 3
(Note, in passing, I:1’s heterozygosity must be inferred from his wife’s and children’s karyotypes. It is a subtle question whether his offspring should properly be included in the analysis, which we will not pursue here). We see that the offspring of heterozygous parents total 14, the proband and the heterozygous antecedents having been excluded. The proportion of abnormal children is 3/14 (0.21). This, then, is a point estimate of the risk for recurrence in a future pregnancy of a heterozygote. The reader should know intuitively that an estimate based on just 14 children is not going to be very precise. And what of children who died in infancy, before the family cytogenetic study has been done? Let us suppose this was the case with III:4 and 5. If there was good evidence for their having been chromosomally abnormal, a better estimate would be 5/14 (0.36).
THE D ERIVATIO N O F A “PRI VAT E ” RECUR R EN CE RIS K F I GU RE
Genetic Heterogeneity and the Use of Empiric Risk Data
We will demonstrate some of the previously noted principles in estimating a private recurrence risk figure for the hypothetical family depicted in Figure 4–1. Six sibships are available for analysis: one in generation II, two in generation III, and three in generation IV. We determine the segregation ratio in each. It is conventional to form a table with a row for each sibship, noting the numbers of phenotypically normal (carrier, noncarrier, unkaryotyped) and
It is not necessarily valid to extrapolate from one family’s experience to a prediction for another. Different factors may cause an abnormality in different families. As an obvious example, it would be misleading to “lump” all Down syndrome families to determine a recurrence risk figure. We need to “split” into the different karyotypic classes of standard trisomy, familial translocations, and de novo translocations. The standard trisomic category Deriving and Using a Risk Figure •
61
FIGURE 4–1 Hypothetical pedigree in which a chromosomal rearrangement is segregating. Filled symbol, abnormal individual with unbalanced karyotype; half-filled symbol, balanced carrier; N in symbol = 46,N. The proband is, as is conventional, indicated by an arrow.
requires further splitting in terms of maternal age. In a unique case, a woman had three trisomy 21 conceptions and displayed a tendency to produce multiple cells with differing (“variegated”) aneuploidies in at least skin, blood, and gonad (Fitzgerald et al., 1986) (and see Chapter 21). She required unique advice. And in reciprocal translocation families, uniqueness is the rule! It is generally reasonable (and often all that is feasible or possible) to apply a risk figure derived from the study of families with a similar, albeit not exactly identical, chromosomal arrangement. But occasionally a family is large enough for a “private” estimate of the recurrence risk to be made from the family itself. This estimate, if it is precise enough (see the later discussion of confidence limits and standard error), is the most valid to offer that family. P REG NA N C Y O UTCO M ES TO WHI C H THE RIS K F IG UR ES RE F E R
With particular reference to the situation of a parent heterozygous for a chromosomal rearrangement, risk figures are generally presented in terms of “the risk that a liveborn child would have a chromosome imbalance related to the parental translocation.” The numerator is the number of aneuploid babies, and the denominator the number of all babies. Thus, considering the example of the common t(11;22)(q23;q11) translocation (p. 86), StengelRutkowski et al. (1988) accumulated data on a total of 318 births (the denominator) to carrier parents, of whom, after ascertainment correction, 9 (the numerator) had the 47,+der(22) aneuploidy; and 62
•
BA SIC CONCEPTS
9/318 gives the risk expressed as a percentage, 2.8%. Separating out mothers and fathers, the respective risk figures are 3.7% (9/241) and <0.7% (0/77). For those choosing prenatal diagnosis, the risk figure of interest relates to the timing of the procedure, generally chorionic villus sampling (usually done at 10–12 weeks) and amniocentesis (15–17 weeks). In other words, they want to know how likely it is they will have to face the actuality of termination. The risk here is likely to be higher (7% in the case of the 11;22 translocation), given that some of the abnormal pregnancies would have spontaneously aborted some time after that period of gestation. Table 4–1 sets out these and other possible ways of considering risk.
Association: Coincidental or Causal? The counselor not infrequently encounters the problem of a chromosomal “abnormality” discovered in a phenotypically abnormal individual, but in whose family others—who are quite normal—are then shown to have, apparently, exactly the same rearrangement. Does a genetic risk apply, then, to children of the carrier, to whom the same rearranged chromosome may be transmitted? From classical cytogenetics, the familial paracentric inversion is a good example. In a review of 69 probands, Price et al. (1987) list the phenotypic abnormalities that led to these individuals coming to a chromosome study. There was a collection of various clinical indications, with no consistent pattern (other than that mental retardation was frequent), and several ascertained quite by chance at prenatal diagnosis.
Table 4–1. Different Ways of Looking at the Quantum of Reproductive Risk due to a Parent Being a Carrier of a Chromosomal Rearrangement NUMERATOR
DENOMINATOR
Abnormal liveborn baby Abnormal liveborn baby
All liveborns All recognized pregnancies Abnormal amniocentesis All pregnancies at result (early second ~16 weeks trimester) 8–14 week miscarriage All recognized pregnancies Abnormal embryo on All embryos from one biopsy IVF procedure Normal embryo All embryos from one on biopsy IVF procedure IVF, in vitro fertilization.
By definition, one parent carries the same inversion; and, if the net is widened, often other relatives as well (Groupe de Cytogénéticiens Français, 1986a). In this context, and provided of course that the carrier relatives are phenotypically normal, one would reach the conclusion that the chromosome rearrangement was balanced, with no functional compromise of the genome; and that it was coincidence that led to its discovery (Romain et al., 1983a). But when some very unusual clinical picture is associated with a paracentric inversion that is rare or previously undescribed (as most inversions are), some writers are skeptical of coincidence and propose a causal link (Urioste et al., 1994a; Fryns et al., 1994). Similarly, Wenger et al. (1995), noting the coincidence of children with an apparently balanced familial translocation, and being phenotypically abnormal, write that “the chance that two rare events in the same individual are unrelated seems unlikely to us.” Here, there is a risk of deception due to “Kouska’s fallacy”—Kouska was a fictional nineteenth-century philosopher who concluded that the combination of unlikely events that led to his parents meeting was too implausible to believe, and that therefore he himself could not exist (Lubinsky, 1986). As does Lubinsky, we must insist on the point: the proband had to be phenotypically abnormal, and the coexistence of a subsequently
discovered different abnormal event (the karyotype) need not be seen as necessarily remarkable. A similar question arises when two rare karyotypes are seen in the same family, or when one individual has more than one aneuploidy. A double aneuploidy such as Klinefelter plus Down syndrome, 48,XXY,+21, could be interpreted as two separately arising nondisjunctions, but each occurring on the basis of the same underlying predisposing factor (such as maternal age). The two conditions occur together more often than the product of the frequency of each singly, which would be consistent with that interpretation. Alternatively, if the XXY component could be shown to reflect a paternal meiotic error, while the trisomy 21 was of maternal origin, then the association could be seen as coincidental. Two different types of abnormality, such as Klinefelter plus Prader-Willi syndrome, a handful of cases of which have been published (Nowaczyk et al., 2004), might also be judged to reflect two unrelated abnormal events, at least for the deletional form of PWS, given that the mechanisms leading to nondisjunction and to deletion are quite different. The prior probability of two abnormal karyotypes coinciding might be a very small figure (1/2000 × 1/15,000 = 1/30,000,000 in the foregoing example); but recalling that the range of abnormal karyotypes is very wide, it should not necessarily be seen as reflecting some extraordinary predisposition when two abnormalities are diagnosed in the one individual or family. Coincidences do happen; and interesting coincidences are publishable (Schneider et al., 2004). In the microarray era, the matter of copy number variants (CNVs) brings the question of causality into a sharp focus, albeit that some of the answers may be less than sharp! As discussed in Chapter 3, a small molecular duplication, for example, that might at first sight seem to be a reasonable candidate as the explanation for a child’s abnormal phenotype, fades in significance when the same observation is made on the DNA sample from a parent. And yet, in the complexity that CNVs present, there may remain a possibility that such a duplication could contribute to abnormality, when existing on a different genetic background. In other words, a particular CNV may be nonpenetrant in a parent, but penetrant in the child; a concept that hitherto has had little relevance in clinical cytogenetics. We can expect that CNV associations, and their causing or not of abnormality, will continue to be an active area of study (and see Chapter 17). Deriving and Using a Risk Figure •
63
Presentation of a Risk Figure A risk figure is a probability statement, and it should be presented as such to the counselee in everyday language—for example: “there is a 50/50 chance for such and such an event”; “the risk for such and such to happen is around 1 chance in 10.” The raw probability figure may not of itself be sufficient, and it is a test of the counselor’s skill to interpret figures so as to provide empathic guidance rather than presumptuous direction. Loaded interpretative comments such as “The risk is quite high that . . .” or “There is only a small chance that . . .” should be used with great care. The perception of a risk figure as high or low may vary greatly according to an individual’s personality and life experiences and the way he or she uses the language of numbers; the very act of discussing the risk may help the client see it in a less threatening light (Kessler and Levine, 1987). Some counselors use diagrams with cartoons showing a crowd of 100 people, with the risk fraction shown in a different color. Dealing with risk advice in a pregnancy, in particular, can be anxiety inducing. Nagle et al. (2009) examined the views of 294 Australian mothers in the postpartum period and recorded preferences for how these women felt, in retrospect, that a risk of having a child with Down syndrome might best have been conveyed. The choices were as follows, with the fractions of the women choosing each category shown in brackets:
(1) As a number in percentage, such as “1%” or “0.05%” (2) In words such as “no increased risk” or “increased risk” (3) As numbers such as “1 in 10” or “1 in 1000” (4) In words such as “high risk” or “low risk” (5) Other (please specify)
[13%]
A combination of the above
[18%]
64
•
BA SIC CONCEPTS
[13%] [37%] [19%] [0%]
None of these stood out as an obvious best, to help the counselor decide on the most appropriate approach. People are different! And people can see to the same risk from different positions. For example, older women having an increased age-related risk (say, 1 in 100) for a child with Down syndrome may decide against an amniocentesis if a maternal serum screening test gives a risk (say, 1 in 200) that is above the cutoff for access to amniocentesis (1 in 250) but lower than their “starting figure”; whereas a younger woman with an age-related risk of, say, 1 in 500 is likely to opt for amniocentesis if she were to have the same 1 in 200 result from the screening test (Beekhuis et al., 1994). PRECI S I O N O F T H E R I S K F I G UR E
As noted earlier, theoretical risk figures are true and empiric risk figures are estimates; the former are exact, and the latter are not. For an empiric figure we have a point estimate (e.g., 10%) and a likely range (e.g., 5%–15%) of where the risk actually is. The more data that have been gathered, the more accurate the estimate and the narrower the likely range; and the more confidently, therefore, can the counselor present the figure. The likely range can be measured in different ways. The standard error, which formally measures the precision of the estimate, can be used to give a sense of the region within which the true risk can realistically be considered to lie. The 95% confidence limits define the broad range that very probably (p = 0.95) encompasses the true risk. Formulae to determine these parameters are set out in Appendix C.
PART TWO PARENT WITH A CHROMOSOMAL ABNORMALITY
This page intentionally left blank
5 autosomal reciprocal translocations
RECIPROCAL TRANSLOCATIONS ARE COMMON, and every counselor can expect to see translocation families. The usual form is the simple, or two-way, reciprocal translocation: only two chromosomes, usually autosomes, are involved, with one breakpoint in each. It is this category we consider in this chapter. The special cases of translocations involving sex chromosomes, and of complex translocations, are dealt with in separate chapters. The translocation heterozygote (carrier) may have a risk to have a child who would be mentally and physically abnormal due to a “segmental aneusomy.” Typically, the imbalance is due to a segment of one of the participating chromosomes being duplicated and a segment of the other chromosome being deleted. This confers a partial trisomy and a concomitant partial monosomy. A few translocations are associated with a high risk, as much as 20%, or very rarely up to 50%, to have an abnormal child. Many translocations imply an intermediate level of risk, in the region of 5%–10%. Some carriers have a low risk, 1% or less; but the woman who is a carrier,
or the partner of a male carrier, may have a high miscarriage rate. Others imply, apparently, no risk to have an abnormal child, but the likelihood of miscarriage is high. Yet others, discovered fortuitously, seem to be of no reproductive significance, with carriers having no difficulties in conceiving or carrying pregnancies and having normal children. The counselor needs to distinguish these different functional categories of translocation, in order to provide each family with tailor-made advice.
BIOLOGY Simple reciprocal translocations arise when a twoway exchange of material takes place between two chromosomes. The process of formation follows the physical apposition of a segment of each chromosome, which may have been promoted by the presence in each segment of a similar DNA sequence. A break occurs in one arm of each chromosome, and the portions of chromosome material distal to the breakpoints switch positions. The distal portions •
67
exchanged are the translocated segments; the rest of the chromosome (which includes the centromere) is the centric segment. The rearranged chromosome is called a derivative (der) chromosome. It is identified according to which centromere it possesses, as in the der(5) and der(10) depicted in Figure 5–1. When no loss or perturbation of genetic material occurs—in other words, the translocation is balanced—the phenotype of the heterozygote is normal, other things being equal. On classical cytogenetics, approximately 1 person in 500 is a reciprocal translocation heterozygote ( Jacobs et al., 1992). The translocation may have arisen de novo in the person, or it may be widespread throughout a family, with many carriers, and sometimes of centuries-long
duration. Koskinen et al. (1993) trace a t(12;21) in western Finland back to a couple born in 1752! When one of the translocated segments is very small and comprises only the telomeric cap of a chromosome arm—and thus we suppose contains no genes—this is regarded as being, effectively, a single-segment exchange. The t(1;4) translocation shown in Figure 5–1, involving a substantial piece of chromosome 4 long arm exchanging positions with the terminal tip of a chromosome 1 long arm, exemplifies single-segment exchange. On the other hand, when both translocated segments are of substantial size, we refer to this as a double-segment1 exchange. The translocation shown in Figure 5–1 between a chromosome 5 and a chromosome 10, with
Translocated segment Centric segments Translocated segment
5
der(5)
der(10)
10
5
der(5)
der(10)
10
der(4)
4
Centric segments Translocated segments
1
der(1)
der(4)
4
1
der(1)
FIGURE 5–1 Reciprocal translocations demonstrating (above) double-segment and (below) single-segment exchange. The translocations are t(5;10)(p13;q23.3) and t(1;4)(q44;q31.3). (Cases of M. A. Leversha and N. A. Monk.)
1 There is scope for confusion in the use of these terms: of course, all reciprocal exchanges, by definition, involve two segments. A true single-segment exchange—that is, a one-way translocation—is generally considered not to exist, in that a segment of chromosome cannot attach to an intact telomere, although there are rare exceptions to this rule. The distinction begins to break down when a translocated segment is very small but could still contain genes (as in the subtelomeric translocation). Be this as it may, the terms doubleand single-segment exchange, used knowledgeably, serve a practical purpose.
68
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
breakpoints in about the mid-short arm of chromosome 5 and a little below the middle of the chromosome 10 long arm, is an example of a double-segment exchange. The translocation involving breakpoints right at, or actually within the centromere, with an exchange of entire arms, is a particular and rare type of double-segment exchange known as a whole-arm translocation (Vázquez-Cárdenas et al., 2007). D E TAILS O F MEIOTI C BE HAV I OR
At meiosis I in the primary gametocyte, the four chromosomes with segments in common come together as a foursome: a quadrivalent. To match homologous segments, the four chromosomes must form a cross-shaped configuration. This is most clearly seen when the chromosomes are at the pachytene stage (Fig. 5–2). As meiosis progresses, the four components of the quadrivalent release their points of attachment except at the tips of the chromosome arms, and they form a ring; if attachment fails, or if one of the terminal pairings release, a chain forms instead of a ring (Oliver-Bonet et al., 2004). With breakdown of the nuclear envelope, spindles forming at each pole of the cell can track to the equator and seek attachment to the centromeres. A cellular motor comes into play, and the chromosome travels to one or other pole. According to which spindle attaches to which centromere—and this may in part be influenced by the configuration of the ring or chain—the distribution of the four homologs to the two daughter gametocytes is determined. Which homologs go to which pole is referred
FIGURE 5–2 Pachytene configuration, simplified outline. The two normal (A, B) and the two translocation (A’, B’) homologs align corresponding segments of chromatin during meiosis I.
to as segregation. The expression 2:2 segregation describes two chromosomes going to one daughter cell, and two to the other. In 3:1 segregation, three chromosomes go to one daughter cell, and one to the other. In 4:0 segregation, all four chromosomes go to one daughter cell, and none to the other. M O D ES O F SEG R EG AT I O N
Within these three broad categories, we can list the particular modes of segregation, according to which chromosomes actually go where. Referring to the four chromosomes of the quadrivalent as A, B, A´, and B´ (Fig. 5–2), the modes of segregation are summarized as follows:
ONE DAUGHTER GAMETOCYTE WITH:
OTHER SEGREGATION DAUGHTER MODE GAMETOCYTE WITH:
2:2 segregations A and B
A´ and B´
A and B´
B and A´
A and A´
B and B´
3:1 segregations A B A´
B´
A B and B´
A´
A´ B´ and A
B
A´ B´ and B
A
4:0 segregation A B A´ B´
none
Alternate segregation Adjacent-1 segregation Adjacent-2 segregation 3:1 segregation with tertiary trisomy or monosomy 3:1 segregation with interchange trisomy or monosomy 4:0 segregation with double trisomy or monosomy
Figure 5–3 depicts five of the possible pairs of daughter gametocytes. Other things being equal, the chromosomal combination is conserved through meiosis II, and the mature gamete forms. From one primary gametocyte, four spermatozoa, or one Autosomal Reciprocal Translocations •
69
FIGURE 5–3 The categories of 2:2 and 3:1 segregation that may occur in gametogenesis in the translocation heterozygote. In the four 3:1 categories, only one of the two possible combinations in each category is depicted (both of each are shown in Fig. 5–4).
ovum and its polar bodies, are thus produced. Gametes from alternate segregation are normal or balanced. Conceptions from adjacent-1 gametes have trisomy for one translocated segment and monosomy for the other. Vice versa, adjacent-2 conceptions have trisomy for one centric segment and monosomy for the other. Tertiary aneuploidies 70
•
have trisomy, or monosomy, with respect to the combined chromosomal content of one of the derivative chromosomes. Interchange aneuploidies have a full autosomal trisomy or a full monosomy. In 4:0 segregation, there is a double trisomy or a double monosomy. Some of the gametes with these unbalanced combinations may be “viable,” in the sense of
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
being “capable of giving rise to a conceptus, which would proceed through to the birth of a child.” Mostly, in fact, they are not. Recombination at meiosis I, and asymmetric segregation at meiosis II, can complicate the story, although this has little practical implication. If recombination occurs in the interstitial segment (between the centromere and the breakpoint), further unbalanced combinations are generated, most of which would not be remotely viable. This phenomenon may possibly have some relevance in preimplantation genetic diagnosis, since testing is done at a stage where there has been little opportunity for selective pressure to have applied. Scriven et al. (1998) list various of these recombination possibilities, and Van Hummelen et al. (1997) diagram the process with respect to a particular translocation on which they had undertaken sperm studies (and illustrate the point that a normal/ balanced gamete can be restored following recombination in adjacent-1 segregation). The most telling evidence that recombination can happen comes from the observation of a meiosis I chromosome having one normal and one derivative chromatid, and polar body analysis has enabled such an observation to be made (Munné et al., 1998a). At meiosis II, asymmetric segregation may lead to two copies of a derivative chromosome being transmitted, as noted later in the section “Meiosis II Nondisjunction.” ALT E R N ATE SEG REGAT I ON
In 2:2 alternate segregation, looking at each centromere in turn around the quadrivalent, one centromere goes to one pole, and the next centromere goes to the other pole. In other words, each centromere goes “alternately”2 to one or the other pole. Thus, the two daughter cells come to contain, respectively, the two normal homologs in one, and the two derivative chromosomes in the other. Note that alternate segregation is essentially the only mode that leads to gametes with a complete genetic complement—one with a normal karyotype, the other with the reciprocal
translocation in the balanced state. All other modes can be classified as malsegregation. ADJACENT SEGREGATION
In 2:2 adjacent segregation, adjacent centromeres travel together (“adjacent” in the sense of centromeres being next to each other, in their positions around the quadrivalent). There are two categories. In adjacent-1 segregation, adjacent chromosomes with unalike (nonhomologous) centromeres travel to the same daughter cell (an aide-mémoire: in adjacent-1, the daughter cells get one of each centromere). Overall, adjacent-1 is the most frequently seen mode of malsegregation in the children of translocation heterozygotes. In adjacent-2 segregation, which is rather uncommon, adjacent chromosomes with like (homologous) centromeres go to the same daughter cell (another aide-mémoire: in adjacent-2, the two homologous centromeres go together). Thus, adjacent-2 segregation rather resembles nondisjunction. 3:1 SEG R EG AT I O N
This is also referred to as 3:1 nondisjunction. Gametes with 24 chromosomes and 22 chromosomes are formed, and the conceptuses therefore have 47 or 45 chromosomes. Almost always, the 47-chromosome conceptus is the only viable one. Two categories exist: either the two normal chromosomes of the quadrivalent plus one of the translocation chromosomes go together to one daughter cell (tertiary trisomy) or, rarely, the two translocation chromosomes and one of the normal chromosomes segregate (interchange trisomy). Tertiary monosomy, with a 45-chromosome conceptus, is extremely rare. Interchange monosomy has never been seen, except at preimplantation genetic diagnosis (PGD). 4:0 SEG R EG AT I O N
In autosomal translocations, 4:0 segregation has been regarded as being of academic interest only. But it may come to have some practical relevance in preimplantation genetic diagnosis. In theory, 16 possible chromosomal combinations could be produced in the gametes of the
2 Not “alternatively,” as some publications erroneously use.
Autosomal Reciprocal Translocations •
71
autosomal translocation heterozygote. Four of these we can, for the most part, ignore (3:1 interchange monosomies and 4:0 segregants), because they are never viable. The two balanced gametes (2:2 alternate segregants) are always viable, other things being equal. Of the remaining 10 possibilities, it is common for none to be viable, with spontaneous abortion the universal outcome. If a translocation heterozygote does have the potential for viable imbalance in an offspring, it is most likely that there will be only one such combination (this was the case in 99% and 100% of translocations, in the considerable experience of two groups; Scriven et al., 1998). Usually, this sole survivable imbalance will be one that endows a partial trisomy. Infrequently two and, very rarely, more than two may be viable. Figure 5–4 depicts the various combinations that may be considered (using the previously discussed t(1;4) translocation as an example). In a review of 1159 translocation families, Cohen et al. (1994) found
the proportions of chromosomally unbalanced offspring as follows: 71% adjacent-1, 4% adjacent-2, 22% tertiary trisomy/monosomy, and 2.5% interchange trisomy. Gamete Studies. It is, apparently, the norm for the heterozygote to produce gametes in which many of the possible chromosomal combinations occur, albeit the proportions may differ, and very substantially so, for some different translocations. Sperm karyotyping results from 44 men, heterozygous for a translocation, are summarized in Table 5–1, along with oöcyte karyotyping data (in most indirectly via polar body analysis) from seven women. Fifty-five percent of sperm, and 70% of ova, were chromosomally unbalanced. The great majority, if not all, of these studied individuals would have presented to the clinic because of reproductive difficulty, and so the data may possibly be biased in the direction of unbalanced forms, compared to the whole
FIGURE 5–4 The full range of segregant gametes that may be produced by the translocation heterozygote, using the t(1;4) depicted in Figure 5–1 as an example. Chromosome 1 chromatin is shown open; chromosome 4 chromatin is crosshatched. 72
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
FIGURE 5–4
(Continued)
population of translocation heterozygotes. On average, alternate and adjacent-1 segregants are the predominant types in spermatogenesis, occurring in fairly similar fractions (43% and 32%, respectively). Adjacent-2 at 13% and 3:1 at 10% are less frequently seen; and barely any 4:0 segregant sperm. Considerable variation occurs: some heterozygotes had no 3:1 segregants, and one had 47%; for adjacent-2, the range is 0% to 40%. The spread of segregant types
seems to be rather similar with men having the same translocation (Benet et al., 2005), such as the related individuals noted in Table 5–1. It would not be surprising if a fairly similar distribution and range of germ cell abnormalities were produced by their heterozygous sisters, although early data from preimplantation diagnosis research do suggest somewhat of a (possibly age-related) propensity in oögenesis for 3:1 segregation (Tables 5–1 and 5–2). Autosomal Reciprocal Translocations •
73
Table 5–1. Chromosome Segregations in Gametes of 51 Male and Female Reciprocal Translocation Heterozygotes, Shown as Percentages in Each Segregant Category t
Male heterozygotes 46,XY,t(1;2)(q32;q36) 46,XY,t(1;4)(p36.2;q31.3)a 46,XY,t(1;4)(p36.2;q31.3)a 46,XY,t(1;9)(q22;q31) 46,XY,t(1;11)(p36.3;q13.1) 46,XY,t(1;13)(q41;q42) 46,XY,t(2;3)(q24;p26) 46,XY,t(2;6)(p12;q24) 46,XY,t(2;9)(q21;p22) 46,XY,t(2;17)(q35;p13) 46,XY,t(2;18)(p21;q11.2) 46,XY,t(3 ;7)(q25;q36) 46,XY,t(3;8)(p13;p21) 46,XY,t(3;11)(q25.3;q25) 46,XY,t(3;15)(q26.2;q26.1) 46,XY,t(3;16)(p23;q24) 46,XY,t(3;19)(p21;p13.3) 46,XY,t(4;6)(q28;p23) 46,XY,t(4;8)(q28;p23) 46,XY,t(4;12)(p11;p11) 46,XY,t(4;17)(q21.3;q23.2) 46,XY,t(5;7)(q13;p15.1) 46,XY,t(5;11)(p13;q23.2)b 46,XY,t(7;14)(q11;q24.1)b 46,XY,t(5;13)(q11;q33) 46,XY,t(5;18)(p15;q21) 46,XY,t(6;9)(p12;q13) 46,XY,t(7;8)(q11.21;cen)a 46,XY,t(7;8)(q11.21;cen)a 46,XY,t(8;9)(q24.2;q32) 46,XY,t(8;22)(q24.22;q11.21) 46,XY,t(9;10)(q11;p11.1) 46,XY,t(9;22)(q21;q11.2) 46,XY,t(10;14)(q24;q32) 46,XY,t(11;17)(q13.1;p11.2) 46,XY,t(11;22)(q23;q11) 46,XY,t(11;22)(q23;q11) 46,XY,t(11;22)(q25;q12) 46,XY,t(14;20)(p11.2;p11.1) 46,XY,t(15;17)(q21;q25)a 46,XY,t(15;17)(q21;q25)a 46,XY,t(15;17)(q21;q25)a 46,XY,t(15;17)(q21;q25)a 46,XY,t(15;22)(q22;q13) 46,XY,t(17;22)(q11;q12) Average fractions* (sperm)
ALT
41 46 39 46 33 41 55 50 43 56 42 28 34 48 48 37 39 46 35 49 57 40 70 30 77 81 24 57 63 44 24 56 56 45 41 22 27 29 51 51 49 48 53 19 19 43%
ADJ-1
ADJ-2
42 6 38 7 50 8 38 13 43 16 42 15 36 7 42 3 28 24 33 11 35 14 46 19 44 21 46 6 36 12 41 16 36 22 52 2 33 20 14 28 35 7 26 17 26 0 48 0 21 2 16 0 14 40 25 11 18 13 41 3 15 19 13 9 26 11 39 12 26 26 14 32 18 13 22 15 19 21 35 9 38 8 40 9 34 11 16 16 13 6 32% 13% Total abnormal = 55%
3:1
4:0
11 9 3 4 8 2 1 4 4 0 8 7 1 0.8 2 5 3 ½ 11 9 2 17 4 17 0 3 22 7 7 9 42 21 6 5 7 30 40 35 4 3 3 2 1.5 43 47 10%
0 0 0 0 0 0 0 0 0 0 0 0 0 0 2 0 0 0 0 1 0 0 0 4 0 0 0 0.04 0.3 0.6 0 0 0.6 0 0 0 0.5 0 0 0 0 0 9 0 0 ~0
Table 5–1. (continued) t
Female heterozygotes 46,XX,t(2;14)(q23;q24) 46,XX,t(2;14)(q31;q24) 46,XX,t(4;14)(p15.3;q24) 4 6,XX,t(6;21)(q13;q22.3) 46,XX,t(7;20)(q22;q11.2) 46,XX,t(9;11)(p24;q12) 46,XX,t(14;18)(q22;q11) Average fractions** (ova)
ALT
11 14 27 0 50 100 40 30%
ADJ-1
ADJ-2
22 22 57 14 55 0 50 0 17 0 0 0 0 60 30% 14% Total abnormal = 70%
3:1
44 14 18 50 33 0 0 26%
4:0
0 0 0 0 0 0 0 0
Notes: The sperm data are arbitrarily set out according to the methodology used. Those down to the t(5;18) were analyzed using the human-hamster hybrid model; the remainder, from the t(6;9), were based upon fluorescence in situ hybridization (FISH) analysis of interphase sperm nuclei (the results from the two approaches are quite similar). The t(7;14) is “out of order”; it is placed beneath the t(5;11) also carried by this subject.b The full dataset according to the two methodologies is contained in Benet et al. (2005), with a total of 89 individuals analyzed. Roundings of percentages have resulted in some data not quite totaling 100%. Some sperm FISH cases were interpreted as showing “other” combinations; these are not listed, and the totals here come to less than 100%. *Sperm: average of the percentages in Tables 1 and 3 in Benet et al. (2005). **Ova: average of pooled oöcyte observations. a Related individuals; note the quite similar fractions. b Both translocations carried by the same man, as a double heterozygote; note the very different proportions from each translocation. Alt, alternate; adj-1, adjacent-1; adj-2, adjacent-2. Sources: Sperm data from Benet et al. (2005). Oöcyte/polar body data, which naturally are based on much smaller numbers (2–11 observations per woman), from Munné et al. (1998b, 1998c), Conn et al. (1999), and Escudero et al. (2000).
Conceptions. It might be expected that the distribution of normal and abnormal conceptions would reflect the distributions of karyotypes in the gametes. Thus, the two men in Table 5–1 with more than 60% alternate segregant forms, they being heterozygous for a t(6;14) and a t(10;12), respectively, might logically be presumed to have a better chance of having a normal child than, for example, the man with a t(1;11) in whose sperm only 33% showed a normal or balanced karyotype. This may be true in some cases, but not necessarily so in all instances, as research from the PGD laboratory is now revealing. Coonen et al. (2000) reported a man with a t(3;11)(q27.3;q24.3), who had 45% alternate forms of segregation on sperm analysis, but of 18 biopsied day-3 cleavage embryos, only three (17%) were normal or balanced. (One of these tested embryos was implanted, and a normal daughter with the balanced translocation subsequently born.) Thus, the fraction of normal/balanced karyotypes fell by one third, from the gametes to the conceptions. His case, as it has turned out, may not be atypical. Initially, in
comparing the pooled segregant distributions of gametes and embryos in each sex, it had appeared that the numbers overall were not grossly discrepant (Table 5–2). However, more recent data published from the ESHRE PGD Consortium have shown that about 80% of embryos from translocation carrier couples are chromosomally abnormal (Goossens et al., 2008). Of 7871 successfully diagnosed embryos, only 1597 (20%) were “transferable,” the fractions nearly identical from male or female carriers. This fraction of 80% is well above the 55% proportion of abnormal sperm (Table 5–1). Very similar data comes from Lim et al. (2008b) and Ko et al. (2010), with ~20% of PGD embryos being normal or balanced; these two groups observed all patterns of malsegregation, including 4:0. Acrocentric chromosomes participating in a reciprocal translocation might be expected to influence segregation, due to the very small lengths of their short arms and thus a marked asymmetry of the quadrivalent (Benet et al., 2005). Lim et al. (2008b) were able to demonstrate the truth of this proposition, in their PGD study mentioned earlier. Autosomal Reciprocal Translocations •
75
Table 5–2. Chromosome Segregations in Embryos of 16 Reciprocal Translocation Heterozygotes Studied at Preimplantation Diagnosis (Shown as Actual Numbers in Each Segregant Category) t
ALT
ADJ-1
ADJ-2
3:1
4:0
Male Heterozygotes 46,XY,t(3;6)(q25;q23) 46,XY,t(3;7)(q23;q36) 46,XY,t(3;7)(q25.3;p22.1) 46,XY,t(11;22)(q23.3;q11.2) Average fractions
3 3 4 9 43%
1 0 1 0 5%
0 0 0 1 2%
Female Heterozygotes 46,XX,t(1;13)(q23;p11) 46,XX,t(1;19)(q32.1;q13.1) 46,XX,t(2;4)(p22.2;q33) 46,XX,t(3;5)(p12;q14.2) 46,XX,t(4;15)(q26;q13) 46,XX,t(5;14)(p15.1;q32.1) 46,XX,t(8;18)(p21.1;q21.1) 46,XX,t(9;20)(q34.2;q11.2) 46,XX,t(11;17)(p15.5;p13) 46,XX,t(11;22)(q23.3;q11.2) 46,XX,t(12;17)(p13;p13) 46,XX,t(14;22)(q11.2;q13.3) Average fractions
7 2 1 3 1 3 4 1 30% 20% Total abnormal = 57%
0 11 2 2 0 4 2 5 9 0 11 6 50%
1 0 1 3 5 0 1 0 0 0 2 0 0 0 3 0 7 0 0 0 4 0 0 3 23% 6% Total abnormal = 50%
1 6 0 0 1 3 0 0 5 2 2 1 20%
0 0 0 0 0 0 0 0 1 1 0 0 2%
Notes: See also Table 25–2. Note that more recent data from ESHRE indicates a higher abnormality rate (about 80%) than the 50%–57% shown here (see text). Alt, alternate; adj-1, adjacent-1; adj-2, adjacent-2. Average fractions are derived from pooling the data in each group. Source: From Mackie Ogilvie and Scriven (2002).
Those translocations that involved an acrocentric chromosome had fewer alternate segregants compared to those that did not (15% cf. 26%), but more 3:1 malsegregants (27% cf. 20%) in 508 embryos analyzed. Viability In Utero. Most unbalanced combinations would produce such enormous genetic imbalance that the conceptus would be lost very early in pregnancy (occult abortion), or even fail to implant. Moderate imbalances would proceed to the stage of recognizable miscarriage or to later fetal death in utero. Only those conceptuses with lesser imbalances might result in the birth of an abnormal child. Viability is much more likely in the case of effective single-segment imbalance, with only one segment of substantial size. In the unbalanced state, a partial monosomy or trisomy for the other very 76
•
small terminal segment is likely to contribute minimally or (if it contains no genes, or at any rate no “dosage vulnerable” genes) not at all to the overall imbalance. This is of particular relevance in adjacent-1 segregation. Consider, for example, gamete (3) in Figure 5–4. The material missing from the telomeric tip of chromosome1 long arm—the telomeric cap—is so small that its loss is, as far as we can tell, of insignificant phenotypic effect. For practical purposes, we can ignore this partial monosomy. So the significant imbalance reduces to a partial 4q trisomy (trisomy 4q31.3-qter). This, as it happens, is well recognized as being a viable complement (and it is the imbalance in the children whose photograph appears in the frontispiece). On the other hand, in the double-segment exchange the imbalance contributed by each segment must be taken into account. Thus, adjacent-1 gametes have both a partial trisomy
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
and a partial monosomy to a significant degree and would produce a “phenotypic hybrid.” Very frequently, the combination is nonviable. If very early miscarriages could be karyotyped, one might expect to discover more of the imbalanced forms. Fritz et al. (2000) conducted such an exercise, using comparative genomic hybridization as the cytogenetic tool. They had identified a family segregating a subtle t(4;12)(q34;p13), in which two children had been born with 46,der(4),t(4;12) (q34;p13), giving a distal 4q monosomy. There had been five previous abortions, and archival pathology material (paraffin-embedded placental tissue) was available for analysis from three of these. A 12- and a 17-week abortus both showed the same karyotype as the surviving children. An 8-week abortus, described as a hydatidiform mole, karyotyped as a tertiary trisomy for almost the whole of chromosome 4: 47,XY,+der(4),t(4;12) (q34;p13), combination (9) in Figure 5–4.
Predicting Segregant Outcomes How can we determine, for the individual translocation carrier, which segregant outcomes, if any, might lead to the birth of an abnormal child? What might be the relative roles of an inherent tendency for a particular type of segregation to occur, and of in utero selection against unbalanced forms? A useful approach is to imagine how the chromosomes come to be distributed during meiosis. Following Jalbert et al. (1980, 1988), we may draw, roughly to scale, a diagram of the presumed pachytene configuration of the quadrivalent, and then deduce which modes of segregation are likely to lead to the formation of gametes, which could then produce a viable conceptus. The following, and with reference to Figure 5–5, are the ground rules: 1. We assume that alternate segregation is (a) frequent and (b) associated with phenotypic normality. 2. The least imbalanced, least monosomic of the imbalanced gametes is the one most likely to produce a viable conceptus.
FIGURE 5–5 Prediction of likely viable segregant outcomes by pachytene diagram drawing and assessment of the configuration of the quadrivalent. Autosomal Reciprocal Translocations •
77
3. If the translocated segments are small in genetic content, adjacent-1 is the most likely type of malsegregation to be capable of giving rise to viable abnormal offspring (Fig. 5–5a). 4. If the centric segments are small in content, adjacent-2 is the most likely segregation to give a viable abnormal outcome (Fig. 5–5b). 5. If one of the whole chromosomes of the quadrivalent is small in content, 3:1 disjunction is the most likely (Fig. 5–5c). The small chromosome may be a small derivative chromosome or a chromosome 13, 18, or 21. 6. If the quadrivalent has characteristics of both Rules 3 and 5, or of Rules 4 and 5, then both adjacent and 3:1 segregations may give rise to viable offspring. 7. If the translocated and centric segments both have large content, no mode of segregation could produce an unbalanced gamete that would lead to a viable offspring (Fig. 5–5d). 8. Subtelomeric translocations may not necessarily form a quadrivalent, and the pairs of homologs might simply join up as bivalents, each pair then segregating independently. Some examples to illustrate these points follow.3 ADJACENT-1 SEGREGATION, S I NG L E-SEG MEN T EXC HA NGE
Many translocations involve an effectively singlesegment exchange, with the “single” translocated segment comprising a fairly small amount of chromatin (1%–2% of the haploid autosomal length, or HAL). This is the classical scenario for adjacent-1 segregation to occur, and to produce a phenotype capable of postnatal survival. The family with the t(1;4) in Figure 5–1, whose children with partial 4q trisomy are shown in the frontispiece as discussed earlier, provides an example. Consider now the family whose pedigree is depicted in Figure 5–6a, in which the individuals shown as heterozygotes have the balanced translocation 46,t(3;11)(p26;q21). A segment of chromatin consisting of almost half of the long arm of chromosome 11, and comprising 1.4% of the HAL, is translocated to the tip of chromosome 3 short arm (Fig. 5–6b). The telomeric tip of chromosome 3 short arm, which we imagine to comprise little or no
phenotypically important genetic material, has moved reciprocally across to chromosome 11. The presumed pachytene configuration during gametogenesis in the heterozygote would be as drawn in Figure 5–6c. The adjacent-1 segregant gamete with der(3) plus normal 11 (heavy arrows) produces a conceptus that has a partial 11q trisomy, since the der(3) carries the segment 11q21-qter. The loss of the 3p telomeric tip in this der(3) we imagine to have no effect. Two, probably three children in the family had been born with this karyotype. No individuals are known having the other adjacent-1 combination (Fig. 5–6c, light arrows), that is to say, the 46,+der(11) karyotype, which would endow a partial 11q monosomy. Consulting Schinzel (2001), viability for the segment 11q21-qter in monosomic state is recorded in only two cases. We assume, therefore, that it has a very high lethality in utero. The scenario of a single survivable imbalanced form, due to a partial trisomy from adjacent-1 segregation in a “single-segment” translocation, as in this t(3;11) example, is, as mentioned earlier, much the most commonly encountered circumstance in translocation families at risk for an abnormal child. Infrequently, both the partial trisomic and the partial monosomic forms are observed. A good example of this is given by distal 4p translocations: both deletion and duplication for this segment are well recognized as having substantial in utero viability. Consider the translocation t(4;12) (p14;p13) described in a family study in Mortimer et al. (1980). A number of family members over three or more generations were balanced carriers, and abnormal children had been born with typical Wolf-Hirschhorn syndrome (all dying in infancy), while others presented the syndrome of partial 4p trisomy (all surviving at least well into childhood). The breakpoints of the translocation are in distal 4p and at the very tip of 12p (12pter). The presumed pachytene configuration would be as drawn in Figure 5–5a (imagining the chromosome 4 chromatin open and chromosome 12 chromatin crosshatched). With such short translocated segments (and very long centric segments), adjacent-1 segregation is the only possibility for viable imbalance. If we ignore the tiny contribution of a duplication or deletion for telomeric 12p—in other words, if we interpret this as an effective single-segment
3 The reader wishing to study further worked examples is referred to Midro et al. (1992), who analyze in some detail a series of translocations of differing risk potentials.
78
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a) N
N
N
N
N
N
(b)
3
der(3)
3
der(3)
der(11)
11
11
3
der(3)
der(11)
11
11
3
der(3)
11
11
(c) 3
der(11)
der(3)
11
FIGURE 5–6 Adjacent-1 segregation. (a) Pedigree of a family in which there segregates a t(3;11)(p26;q21) having the characteristics associated with adjacent-1 malsegregation. Two independently ascertained probands have a partial 11q trisomy, and a deceased relative, who died at age 18 in an institution for the retarded, had a similar appearance from photographs, and so very probably had the same karyotype. Filled symbol, unbalanced karyotype; half-filled symbol, balanced carrier; N in symbol = 46,N; small diamond, prenatal diagnosis; arrow, proband. (b) Partial karyotype of a translocation heterozygote (above), showing the 3;11 translocation, and a child with the unbalanced complement (below) (case of A. J. Watt). (c) The presumed pachytene configuration during gametogenesis in the heterozygote (chromosome 3 chromatin, open; chromosome 11 chromatin, crosshatched). Arrows indicate movements of chromosomes to daughter cells in adjacent-1 segregation; heavy arrows show the combination observed in this family.
imbalance—the situation reduces to the two possible adjacent-1 outcomes being a partial 4p trisomy and a partial 4p monosomy. Both of these are recognized entities, as noted earlier, and apparently both have substantial viability in utero. The abnormal karyotypes would be written 46,der(12)t(4;12) (p14;p13) and 46,der(4)t(4;12)(p14;p13). If the “other segment” comprises acrocentric short arm, then, given the redundancy of this material, the case for considering the translocation as a single-segment entity is particularly valid, with the resulting imbalances being “pure.” De Carvalho et al. (2008) illustrate this in their report of a large family segregating a t(5;15)(p13.3;p12) translocation, in which the extraordinary total of 21 individuals were known with either a deletion or a duplication of distal 5p. Applying the principles set out later in this chapter, the risk to the carrier in this family to have a child with an imbalance—presumably due to adjacent-1 malsegregation—is very high, at 54%. Essentially, as it would seem, alternate and adjacent-1 segregant outcomes were equally probable. ADJACENT-1 SEGREGATION, D OU B LE-SEG MEN T E XC HA NGE
With a double-segment translocation, an adjacent-1 imbalanced conceptus has both a partial trisomy and a partial monosomy (also called a duplication/ deficiency, or duplication/deletion, abbreviated to dup/del). The combined effect of the two imbalances is more severe than either separately. Thus, it is infrequent that the carrier of a “double-segment” exchange can ever have a chromosomally unbalanced pregnancy proceeding through to term, or close to term. Multiple miscarriage is the typical observation (e. g., Fig. 5–16). But occasionally viability is observed for one, or rarely both, of the dup/ del combinations. Nucaro et al. (2008) studied a t(3;10)(p26;p12) family with affected individuals in three generations, and yet all still living, and able to be examined and their karyotypes confirmed as 46,der(3)t(3;10)(p26;p12), conveying a partial 3p monosomy and 10p trisomy; the countertype adjacent-1 karyotype was not observed, but it may well have been the cause of the several miscarriages
recorded. The double-segment t(4;8)(p16.1;p23.1) depicted in Figure 5–7 has very small translocated segments: the tip of chromosome 4 and the tip of chromosome 8 have exchanged positions.4 In this family, each of the two possible adjacent-1 segregant outcomes were observed: the index case with del(4p)/dup(8p), and his uncle with dup(4p)/ del(8p). In the former, a Wolf-Hirschhorn gestalt was discernible, reflecting the del(4p) component. A similar example is seen in the family reported in Rogers et al. (1997). They provide in their paper a photograph of six siblings sitting on a sofa in 1958, one with a dup(11q)/del(4q) karyotype, two who since died presumed to have been del(11q)/ (dup(4q), and one girl carrying the family t(4;11) (q34.3;q23.1) who went on to have, in the next generation, a del(11q)/(dup(4q) child. Exceptionally, both translocated segments can be of substantial size and yet be survivable, if barely, to term. The outlying points in Figure 5–17 reflect such cases. The double-segment t(5;10)(p13;q23.3) exchange illustrated in Figure 5–1 provides an example, this translocation having been identified in a family following the death of a neonate with multiple malformations. The genetic abnormality comprises a deletion of 5p and a duplication of 10q, for a total imbalance of 2.5% HAL (1.1% HAL monosomy plus 1.4% HAL trisomy). When entire arms of chromosomes are translocated (whole-arm translocation), it is almost always so that the unbalanced segregants would be unviable (Vázquez-Cárdenas et al., 2007). Rare exceptions exist. Czakó et al. (2002) report a t(18;20) (p11.1;p11.1), in which the abnormal child of a carrier father was effectively trisomic for all of 20p and monosomic for all of 18p (1.0% HAL trisomy plus 0.8% HAL monosomy). The woman with a wholearm 15p;16q translocation described in Chen et al. (2004d) had a history of miscarriage and stillbirth, and two further pregnancy losses proven to have complete 16q trisomy, this imbalance conveying as much as 2.1% HAL trisomy, and associated with a recognized severe phenotype. (Since the concomitant 15p monosomy presumably did not contribute to the fetal defects, this example has more of the flavor of a single-segment translocation.)
4 This same 4;8 translocation has been observed in a small number of unrelated families, and it may be, after the t(11;22) noted below, the most frequent human reciprocal translocation. This recurrence reflects the presence in distal 4p and 8p of “olfactory-receptor clusters,” which can act as recombination-predisposing duplicons (Maas et al., 2007). Other recurrent rearrangements are the translocations t(4;18)(q35;q23) and t(8;22)(q24.13;q11.21) (Horbinski et al., 2008; Sheridan et al., 2010). Some apparent recurrences may actually reflect unrecognized identity by descent (Youings et al., 2004).
80
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
FIGURE 5–7 Adjacent-1 segregation, double-segment translocation with very small segments. (a) Parent with the translocation t(4;8)(p16.1;p23.1). The index case, his child, has the karyotype 46,+der(4) and so has a del(4p)/dup(8p) imbalance, and an uncle has the countertype dup(4p)/del(8p) imbalance due to the 46,+der(8) karyotype. (Case of C. E. Vaux.) (b) The presumed pachytene configuration during gametogenesis in the heterozygote (chromosome 4 chromatin, open; chromosome 8 chromatin, crosshatched). Arrows indicate movements of chromosomes to daughter cells in adjacent-1 segregation. The upper combination (light arrows) would produce the dup(4p)/del(8p) imbalance, and the lower (heavy arrows) the del(4p)/dup(8p) imbalance.
The opportunity occasionally arises to provide direct evidence of early in utero lethality of a particular imbalanced state. In a family study of a t(8;18) (p21.3;p11.23), Cockwell et al. (1996b) demonstrated on a severely malformed spontaneously aborted 11-week fetus one of the adjacent-1 conceptions, the dup(8p)/del(18p) state. This chromosomal constitution caused a double-segment imbalance, with a trisomy for 8p21.3-pter, and a monosomy for 18p11.23-pter, giving a combined 1.2% HAL imbalance (0.8% for trisomy, 0.4% for monosomy).
The countertype dup(18p)/del(8p) karyotype had produced, in this family, a child with an abnormal phenotype. Atypically, this viable form had more HAL monosomy than trisomy. ADJACENT-1 SEGREGATION WITH SU BT ELO M ER I C D O UB L E-SEG M EN T E XC H A N G E
With subtelomeric fluorescence in situ hybridization (FISH) or microarray, translocations involving Autosomal Reciprocal Translocations •
81
submicroscopic (typically <2–3 Mb) segments in the (generally) gene-rich regions just below the telomeres can be identified. Consider the family reported in Vogels et al. (2000), the pedigree of which is presented in Figure 5–8a. Three individuals (two cousins and the nephew of one of them) presented a similar picture of severe mental defect and “obnoxious” behavior, with facial dysmorphism and certain malformations; another with similar facial dysmorphism had died of a heart defect as a neonate. This picture demanded close attention to the possibility of a chromosomal explanation, but highresolution karyotyping was normal in all four, with one of the cousins having been tested on several occasions. Finally, FISH with subtelomeric probes provided a clear and unambiguous illumination (Fig. 5–8b). Testing the potential carriers in the family, a 5q subtelomeric probe and an 18q subtelomeric probe hybridized properly to one chromosome 5qter and to one chromosome 18qter; but the other 5qter hybridized with the 18q probe, and the other 18qter carried the 5q probe. Thus was demonstrated a double-segment reciprocal exchange between 5q and 18q: 46,t(5;18)(subqter;subqter).5 The affected individuals were trisomic for a very small segment of subterminal 5q, and monosomic for a similarly small segment of 18qter, and the clinical picture suggested features of both the dup(5q) and the del(18q) syndromes. A number of similar families are now being reported, and many genetics services are reviewing their “chromosomal-seeming families” in which classical cytogenetics had failed to reveal any abnormality. A recurring theme in microarray analysis is the recognition of a terminal deletion accompanied by duplication of terminal material from another chromosome (e.g., Izumi et al., 2010); and when probes for the specific regions are “FISHed back” to a metaphase spread, the duplicated segment can be seen as translocated to the deletion site. Many will be de novo, but a proportion are the consequence of a balanced parental reciprocal translocation. If a quadrivalent were to form in a subtelomeric translocation, practically always it would only ever be the adjacent-1 gametes that would be viable, besides the normal and balanced forms. Furthermore, it is unlikely that any other segregation pattern than 2:2 would occur. With such tiny chromosomal segments involved, it is probable that the homologs, the
normal and the derivative, would simply pair up as in a normal bivalent, leaving the tiny nonhomologous segments at the ends unpaired. In that case, the expected segregations at meiosis would be random, with equal probability for each outcome, namely, normal:balanced:(dup/del):(del/dup) in the ratio 1:1:1:1. ADJACENT-2 SEGREGATION
This is an uncommonly observed mode of segregation, typically limited to translocations in which the two participating chromosomes each has a short arm of small genetic content, and small enough that the whole short arm can be viable in the trisomic state. In fact, most cases involve an exchange between chromosome 9 and an acrocentric, or between two acrocentrics (Duckett and Roberts, 1981; Stene and Stengel-Rutkowski, 1988; Chen et al., 2005d). The breakpoints characteristically occur in the upper long arm of one chromosome and immediately below the centromere in the long arm of the other (an acrocentric). Thus, the centric segments are small. The t(9;21)(q12;q11) illustrated in Figure 5–9a exemplifies the adjacent-2 scenario. At meiosis I, the form of the quadrivalent would be as drawn in Figure 5–9b. The “least imbalanced, least monosomic” gamete from 2:2 malsegregation is the one receiving chromosome 9 and the der(9) (heavy arrows). The conceptus will have, in consequence, a duplication of 9p (and a small amount of 9q heterochromatin) and a deletion of 21p (and a minuscule amount of subcentromeric 21q). Although comprising a substantial piece of chromatin (1.8% of HAL), 9p is qualitatively “small” in the trisomic state. Monosomy for 21p is without effect, and the 21q loss makes little if any contribution, and thus the picture is practically that of a pure 9p trisomy. This is a known viable aneuploidy. The countertype gamete with the der(21) causes monosomy 9p and is not viable. A very similar circumstance applies with the t(4;13)(q12;q12) described in Velagaleti et al (2001); the open and crosshatched chromosomes in the cartoon karyotype (Fig. 5–9) could be regarded, for this example, as chromosomes 4 and 13, respectively. The index case in this family was trisomic for all of 4p, and the small segment 4cen-q12 (and monosomic for the tiny segment 13p-q12), having the karyotype 46,XY,+der(4),–13.
5 This is not official ISCN nomenclature.
82
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
(c)
FIGURE 5–8 The subtelomeric translocation. (a) Pedigree of family. Filled symbol, abnormal individual with subtelomeric aneusomy; half-filled symbol, balanced carrier; dot, miscarriage. (From Vogels et al., 2000; courtesy J.- P. Fryns.) (b) Fluorescence in situ hybridization (FISH) demonstration of a reciprocal translocation t(5;18), undetectable on high-resolution G-banding (middle), but obvious using probes for the 5q subtelomeric region (left), and the 18q subtelomeric region (right). (c) Segregation patterns at meiosis I in the heterozygote, making the assumption, which may or may not be correct, that the homologs align as independent bivalents, rather than as a quadrivalent. Segregation would thus be 1:1 for each pair of bivalents. If the pairs of bivalents happen to be disposed on either side of the equatorial plate as in (c) upper, normal and balanced daughter cells are produced. If they happen to be disposed as in (c) lower, unbalanced cells result, either with del(5q)/ dup(18q) (thin arrows), or, as observed in this family, dup(5q)/del(18q) (thick arrows). The equatorial plate, in transverse section, is indicated by the dotted line. This scenario will commonly apply to the submicroscopic reciprocal translocation identified following microarray analysis on an index case.
(a)
9
9
der(9) der(21) 21
9
der(9)
21
9
der(9)
der(21)
21
9
9
der(9)
21
(b)
9
der(21)
der(9)
21
FIGURE 5–9 Adjacent-2 segregation. (a) Mother (above) has a reciprocal translocation t(9;21)(q12;q11), and her child (below) has the adjacent-2 karyotype 46,+der(9)t(9;21)(q12;q11). (Case of C. M. Morris and P. H. Fitzgerald). (b) The presumed pachytene configuration during gametogenesis in the heterozygote (chromosome 9 chromatin, open; chromosome 21 chromatin, crosshatched). Arrows indicate movements of chromosomes to daughter cells in adjacent-2 segregation; heavy arrows show the viable combination, as observed in this family.
The del(22)(q11) syndrome, so well known otherwise due to microdeletion, can also arise from a familial translocation, and this provides an example of a double-segment imbalance with adjacent-2 segregation. Imagine a t(9;22)(q12;q11.21) with the 22q breakpoint just below the DiGeorge critical region (DGCR). Considering the crosshatched chromosome in Figure 5–9b to be a chromosome 22, then the der(9) will lack the DGCR. A 46,+der(9),–22 child from adjacent-2 segregation (the heavy arrows) will have the 22q deletion syndrome, superadded upon a 9p trisomy. Pivnick et al. (1990) and El-Fouly et al. (1991) describe children 84
•
in whom these separate-and-together dup(9p) and del(22q) phenotypes could be distinguished. A double-segment exchange with both adjacent-2 segregants observed, and reflecting a parentof-origin effect, is shown in the family reported by Abeliovich et al. (1996). The family translocation, carried by the father, was due to breakpoints in the long arms of chromosomes 15 and 21, t(15;21) (q15;q22.1). Both centric segments, 15pter-15q15 and 21pter-21q22.1, are of quite substantial size. One child had the karyotype 46,–15,+der(21), with a proximal partial 15q monosomy and a proximal partial 21q trisomy. The phenotype was predominantly
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
that of the Prader-Willi syndrome (PWS), reflecting the lack of a paternally contributed PWS critical region, residing in 15q11-q13. There was no clearly apparent contribution from the partial trisomy for 21pter-21q22.1. The other child, with a dup(15q)/ del(21q) combination, 46,+der(15),–21, displayed a combination of features due to monosomy 21pter21q22.1 and trisomy 15pter-15q15. An analogous story is that of a mother carrying a translocation t(15;22)(q13;q11.2), and in this case her child with the 46,–15,+der(22) combination presented the clinical picture of Angelman syndrome (AS), due to absence of a maternally originating AS critical region in 15q11–13 (Kosaki et al., 2009). Another child of hers had the opposite adjacent-2 imbalance, 46,–22,+der(15), and his phenotype was that of DiGeorge syndrome. A double-segment case in which the two centric segments were much smaller is exemplified in Chen et al. (2005d). Here, in a 14;21 rearrangement, described as t(14;21)(q11.2;q11.2), both breakpoints were in the first subband below the centromere. The der(14) thus comprised almost all chromosome 21 material, with just the short arm, centromere, and a very small amount of proximal long arm being from chromosome 14; and vice versa, the der(21) consisted largely of chromosome 14 material. Three affected family members, two
brothers and their aunt, carried the der(14) in unbalanced state due to adjacent-2 segregation and were thus trisomic for the small proximal 14q segment and monosomic for the small proximal 21q segment. The dysmorphology was quite mild, but the functional neurobehavioral phenotype was rather severe. A very similar (and possibly identical) scenario is described in Dave et al. (2009): the carriers in this family typed 46, t(14;21)(q21.2;q21.2), and the three affected individuals as 46,XX,+der(14) t(14;21)(q21.2;q21.2),–21. This sort of translocation, with very proximal q arm breakpoints in acrocentric chromosomes, may require microarray, with FISH confirmation, for its recognition, as Koochek et al. (2006) show in a t(14;15)(q11.2.;q13.3). Affected individuals inheriting a duplication of proximal 15q due to a maternal adjacent-2 malsegregation displayed a phenotype of which autism was a prominent feature. The reason so few examples of adjacent-2 segregants are seen is that most convey a lethal imbalance during early embryogenesis. Naturally, if the window of observation were to be shifted to this period of development, more cases would reveal themselves. An example is shown in Figure 5–10, this being the karyotype from the products of conception obtained at miscarriage in the first trimester from a woman who was herself a translocation
FIGURE 5–10 Adjacent-2 segregation, with an imbalance lethal in early pregnancy. The mother (above) has the karyotype 46,XX,t(13;16)(q12.3;q13). Tissue from the products of conception of a spontaneous firsttrimester abortion was cultured, and the chromosomal complement from these cells (below) showed the karyotype 46,XX,–13,+der(16). There is monosomy of proximal 13q for a segment of HAL 0.6%, and partial trisomy 16 for a segment of HAL 2.1%. (Case of M. D. Pertile.) Autosomal Reciprocal Translocations •
85
carrier, 46,XX,t(13;16)(q12.3;q13). The karyotype of the cultured products, 46,XX,–13,+der(16), displays an overall HAL imbalance of 2.6%. Two previous miscarriages to this couple might possibly also have had this karyotype. Earlier in the piece, at the 3-day embryo stage, selection pressures have not yet come to bear; and thus three embryos at preimplantation diagnosis, all with an adjacent-2 imbalance, as seen from a 46,XY,t(10;18)(q24.1;p11.2) carrier, is perhaps not too remarkable a finding (Munné et al., 2000b). 3:1 SEG R EG ATIO N WI T H TE RT I A RY TR IS O MY
Tertiary trisomy is fairly uncommon—or to be precise, fairly uncommonly seen in a term pregnancy— and may arise only when one of the derivative chromosomes is of small content. It exists in the abnormal individual as a supernumerary chromosome, with the karyotype 47,+der. The centric segment will necessarily contain the whole short arm of the derivative chromosome, and it will necessarily be of a chromosome having a small short arm. Almost always, complete long arms (and in fact most complete short arms) contain too much material to allow viability in a supernumerary derivative chromosome, and spontaneous abortion ensues. A rare chance to illustrate this point is given in Fritz et al. (2000) who, as mentioned earlier, studied archived material from an abortus, the mother carrying a subtle translocation, 46,XX,t(4;12)(q34;p13). They showed (by CGH) a tertiary trisomy, 47,XY,+der(4), with almost the entire chromosome 4, and the tip of 12p, present as an additional chromosome. There is, as noted later, a significant maternal age effect in 3:1 imbalance. Curiously enough, in the most common, by far, human reciprocal translocation, practically all abnormal offspring of the heterozygote have a tertiary trisomy, due to 3:1 meiosis I malsegregation. This is the t(11;22)(q23;q11)6 (Fig. 5–11a). The quadrivalent of this 11;22 translocation would have the form outlined in Figure 5–11b. The content of
the smallest chromosome, the der(22), is small (respecting the requirement for the derivative to have a small short arm, chromosome 22 easily qualifies), and its major genetic composition is accounted for by the distal 11q segment. The presence of this 47th chromosome does not necessarily impose a lethal distortion on intrauterine development, and a pregnancy could continue through to the birth of a child who would have trisomy for the segment 11q23-qter (and for the very small segment 22pterq11), with the karyotype 47,+der(22),t(11;22) (q23;q11). Carter et al. (2009) review the clinical features associated with this imbalance, now known as Emanuel syndrome. This t(11;22) is the spectacular exception to the rule that, in different families, translocations arise at different sites. The great majority of families have a “private translocation,” and many may represent the first and only case in the whole of human evolution. Apparently, few predispositions for specific rearrangement exist; equally apparently, 11q23 and 22q11 show a remarkable predisposition, which may reflect a physical proximity between the two chromosomes during meiosis (Ashley et al., 2006). Kurahashi and Emanuel (2001) studied normal volunteers, and, being able to test very large numbers of sperm, they could show that de novo t(11;22) (q23;q11) translocations must be being generated from time to time; and Ohye et al. (2010), studying eight de novo cases, showed the translocation in each to have been of paternal origin. The male t(11;22) heterozygote, at least, does produce other types of unbalanced gamete, as shown on sperm chromosome study (Table 5–1), but none of these is ever viable.7 Using FISH, Estop et al. (1999) showed in one subject 3:1 segregation in 40% of sperm, and they note a predisposition of this translocation to segregate according to this mode. Data from embryos tested at PGD from t(11;22) carriers are set out in Table 26–1, and these show a much lower frequency of 3:1 embryos from the male carrier (2%), compared with the female (30%), and compared with the 40% sperm rate just mentioned.
6 One breakpoint may compromise the function of a tumor suppressor gene; there is preliminary evidence that the t(11;22) carrier has an increased risk for breast cancer (Lindblom et al. 1994), but a definitive study, looking at large numbers in several kindreds, has yet to be done. 7 Except in the extraordinary setting of postzygotic rescue. Kulharya et al. (2002) report a t(11;22) carrier mother having had a child from presumed adjacent-1 segregation with 46,XY,der(22) at conception, and then mitotic loss of the der(22) in one cell and duplication of the normal 22, leading to 46,XY,der(22)/46,XY mosaicism.
86
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
FIGURE 5–11 Tertiary trisomy. (a) The common t(11;22)(q23;q11) in the heterozygous state (above) and in the typical unbalanced state (below). (b) The presumed pachytene configuration during gametogenesis in the heterozygote (chromosome 11 chromatin, open; chromosome 22 chromatin, crosshatched). Arrows indicate movements of chromosomes to daughter cells in a 3:1 tertiary segregation; heavy arrows show the viable trisomic combination.
Note the point that probands in whom a supernumerary marker chromosome (SMC) is discovered are often found, on parental study, to have a derivative chromosome reflecting a tertiary trisomy (Stamberg and Thomas, 1986). Braddock et al. (2000) describe a family in which an SMC due to 3:1 malsegregation had, initially, escaped recognition as such. A child with “atypical Down syndrome” had been karyotyped as trisomy 21. On attending a Down syndrome clinic at age 9 years, the clinical picture raised doubt and his chromosomes were restudied. He turned out to have a tertiary trisomy for a der(21), which comprised much of chromosome 21 and a small part of distal 5p. His mother and several other relatives carried a t(5;21) (p15.1;q22.1), and a similarly abnormal aunt had the same tertiary trisomy, 47,+der(21). This story has lessons both for cytogeneticists and genetic counselors.
3:1 SEG R EG AT I O N W I T H T E RT I A R Y M O N OS O MY
If one derivative is very small, and the amount of material that is missing is “monosomically small,” the countertype 3:1 22-chromosome gamete may lead to a viable conceptus. Consider the 12;13 translocation t(12;13)(p13.32;q12.11) shown in Figure 5–12a. The large derivative chromosome is not far from being a composite of the two complete chromosomes. It is missing only subterminal 12p and pericentromeric chromosome 13. This is a “small” loss, and thus the 45,der(12) conceptus is viable (Fig. 5–12b). Any initially 45-count karyotype obliges consideration that there may, in fact, be a tertiary monosomy. For example, Courtens et al. (1994) describe an infant who died at birth with, at first sight cytogenetically, monosomy 21 (45,–21). But with FISH Autosomal Reciprocal Translocations •
87
(a)
(b)
FIGURE 5–12 Tertiary monosomy. (a) Mother (above) has a reciprocal translocation between nos. 12 and 13, 46,t(12;13)(p13.32;q12.11). Two children (below) inherited the derivative 12, but no normal chromosome 12 or 13 from the mother, and have the karyotype 45,der(12). They are thus monosomic for the tip of 12p and pericentromeric 13 (and only a mildly abnormal phenotype). Chorionic villus sampling in a subsequent pregnancy gave a 46,XX result; an elder sister was a balanced carrier. (Case of M. D. Pertile.) (b) The presumed pachytene configuration during gametogenesis in the heterozygote (chromosome 12 chromatin, open; chromosome 13 chromatin, crosshatched). Arrows indicate movements of chromosomes to daughter cells in a 3:1 tertiary segregation; heavy arrow shows the monosomic complement. Alternatively, the three large chromosomes might form a trivalent, and the tiny der(13), being unattached, might segregate at random.
and molecular studies, a 45,+der(1) from a maternal 1;21 translocation was discovered. Sometimes the two phenotypes of the two contributing monosomies can be separately discerned. Thus, Reddy et al. (1996) describe children with a combined Di George (DGS) and Wolf-Hirschhorn (WHS) phenotype, having the karyotype 45,der(4) t(4;22)(p16.3;q11.2)mat. The large derivative chromosome comprised almost all of 4 and almost 88
•
all of 22q, but it lacked the WHS and DGS critical segments. Similarly, McGoey and Lacassie (2009) give an account of the child of a carrier father who had features of both DGS and subtelomeric 9q deletion, with the karyotype 45,XX,der(9)t(9;22) (q34.3;q11.2)pat. Wenger et al. (1997) report a mother with a t(8;15)(p23.3;q13) whose child had the karyotype 45,der(8) and presented a phenotype with features of Angelman syndrome (due to loss of
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
the maternally originating segment 15q11-q13) and of 8p–syndrome. Torisu et al. (2004) describe a severely retarded, epileptic child with tertiary monosomy dictating a combination of Angelman syndrome and the 1p36 deletion syndrome: her karyotype was 45,XX,der(1)t(1;15)(p36.31;q13.1) mat. An interesting historical example, in that it provided a key observation toward the discovery of the TSC2 locus, is that of a child with 45,der(16), who had monosomy for the segment 16p13-pter, and who had both tuberous sclerosis and polycystic kidney disease, due to loss and disruption, respectively, of the adjacent TSC2 and PKD1 loci. The heterozygous 46,t(16;22) family members had polycystic kidney disease, due to the disruption of PKD1 (European Polycystic Kidney Disease Consortium, 1994). However, the great majority of conceptions with a tertiary monosomy are expected to be lethal in utero. A direct demonstration of this circumstance is illustrated in the case of a 3:1 malsegregation of a maternal t(11;22) in a spontaneous abortus at 7 weeks gestation with 45,der(11), which resulted in monosomy for distal 11q and monosomy for proximal 22q ( Jobanputra et al., 2005). 3:1 SEG R EG ATIO N WI T H I NT E R C H A N G E TRIS OMY
This mode of segregation can only produce a liveborn child when a “trisomically viable chromosome” (i.e., 13, 18, or 21, or even 22) participates in the translocation (Fig. 5–13a). This chromosome accompanies the two translocation (interchange) elements of the quadrivalent to one daughter cell (Fig. 5–13b). Interchange trisomy 21 is rare, interchange trisomies 13 and 18 extremely rare, and interchange trisomy 22 barely recorded (Stene and Stengel-Rutkowski, 1988; Teshima et al. 1992b; Koskinen et al., 1993; Patel and Madon, 2004). Concerning other (nonviable) autosomes, there are examples of interchange trisomies 2, 6, and 7, from a translocation parent, these aneuploidies having been identified at PGD, or upon analysis of abortus material (Cockwell et al., 1996a; Conn et al., 1999; Lorda-Sánchez et al., 2005). Theoretically, uniparental disomy can be a consequence of interchange trisomy, if one of the “trisomic” chromosomes is subsequently lost postzygotically, and if this chromosome had come from the noncarrier parent. If this
chromosome is one that is subject to imprinting according to parent of origin, phenotypic abnormality will be the consequence, notwithstanding the apparently balanced karyotype, the same as the parent’s. Thus, for example, a 46,t(8;15) father could have a 46,t(8;15) child with Angelman syndrome, or a mother a child with Prader-Willi syndrome. Actual examples of this type of mechanism are extremely rare (Kotzot, 2001; Dupont et al., 2002; Heidemann et al., 2010).
3:1 SEG R EG AT I O N W I T H I N T ER CH A N G E M O N OS O MY
Autosomal monosomy is typically associated with very early arrested development of the embryo, by the blastocyst (day 5) stage. Only with PGD does a practical relevance possibly emerge, since there has not yet been the chance for selection pressure to have operated. In the PGD case reported in Conn et al. (1999) noted earlier, the woman being a t(6;21) heterozygote, a transferred embryo that implanted only transiently may have had an interchange monosomy 6. Yet to be observed is uniparental disomy following “correction” by duplication of the single normal homolog in the embryo resulting from interchange monosomy. The countertype gamete in Figure 5–13a, for example, would be nullisomic for 21. Replication of the chromosome 21 from the other gamete could restore disomy and with a normal karyotype. Note that this would be uniparental isodisomy and from the other parent. 4:0 SEG R EG AT I O N
A total nondisjunction of the quadrivalent complex is rare indeed. FISH allows for very large numbers of sperm to be analyzed, and so, in a few cases, a very few (fractions of a percent) 4:0 gametes are seen (Table 5–1). If 4:0 segregation should happen, preimplantation lethality would, in most, be the likely consequence. Out of interest, the reader may care to note how a hypothetical double trisomy of 18 plus 21, based on the 4:0 combination in Figure 5–4 (15), and potentially associated with some in utero survival (Reddy, 1997), could come from the t(18;21) shown in Figure 5–15. PGD has now brought the 4:0 gamete out from its former place of practical irrelevance. Autosomal Reciprocal Translocations •
89
(a)
(b)
FIGURE 5–13 Interchange trisomy. (a) Mother (above) has a reciprocal translocation between nos. 12 and 21; her child (below) inherited the maternal translocation chromosomes and a “free” chromosome 21. The breakpoints are 12q13.1 and 21p13; an apparent gap, comprising satellite stalk, can be discerned between the centromere of the der(21) and its 12q component. (Case of R. Oertel.) (b) The presumed pachytene configuration during gametogenesis in the heterozygote (chromosome 12 chromatin, open; chromosome 21 chromatin, crosshatched). Arrows indicate movements of chromosomes to daughter cells in 3:1 interchange segregation; heavy arrows show the trisomic combination.
MORE THAN ONE UNBALANCED SEG REG A N T TY P E
Sometimes a reciprocal translocation has characteristics associated with more than one type of malsegregation; so each type may be seen in the family (Abeliovich et al., 1982). Consider the 11;18 translocation t(11;18)(p15;q11) shown in Figure 5–14. First, the translocated segments are small: 18q is 90
•
known to be viable in the trisomic state, and the tip of 11p contributes a minimal/nil imbalance (i.e., this is regarded as a single-segment imbalance). Thus, one of the adjacent-1 segregants is presumed to be viable. Second, two component chromosomes of the pachytene configuration, the der(18) and chromosome 18, are of small overall genetic content. Thus, 3:1 segregation with either tertiary trisomy or interchange trisomy is possible. In the
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
(c)
FIGURE 5–14 More than one viable segregant form. (a) Pedigree. Filled symbols, unbalanced karyotype, as shown; half-filled symbols, heterozygote. (b) Mother and one daughter have a reciprocal translocation of chromosomes 11 and 18, t(11;18)(p15;q11) (upper). Each had one unbalanced offspring, one having 47,+der(18) due to 3:1 tertiary trisomy (middle), and the other 46,+der(11) from adjacent-1 segregation (lower). The former had a complete trisomy 18p and the latter a partial 18q trisomy. (Case of C. Ho and I. Teshima.) (c) The presumed pachytene configuration during gametogenesis in the heterozygote (chromosome 11 chromatin, open; chromosome 18 chromatin, crosshatched). Heavy arrows indicate one adjacent-1 segregant movement of chromosomes, and light arrows indicate movements of chromosomes to daughter cells in a 3:1 tertiary trisomy segregation, each of which occurred in this family. (From Gardner et al., 1978.)
event, the two unbalanced karyotypes in this family reflected adjacent-1 and 3:1 tertiary trisomy segregation. The t(9;21) discussed earlier as an example of adjacent-2 segregation could also, in theory, produce a second viable complement, interchange trisomy 21. Rather more spectacular is the translocation illustrated in Figure 5–15. A mother had the karyotype 46,XX,t(18;21)(q22.1;q11.2): these breakpoints are toward the end of 18q and immediately below the centromere in 21q. She had a stillborn child with tertiary monosomy, a miscarriage with adjacent-1 malsegregation (and two other unkaryotyped miscarriages), and a surviving child with tertiary trisomy. These three karyotyped pregnancy
outcomes were, respectively, 45,der(18), 46,der(18), and 47,+der(18). An uncle said to have had Down syndrome may have had the 46,der(18) karyotype (the der(18) includes the segment of 21 that contributes substantially to the DS phenotype), or possibly interchange trisomy with 47,+21,t(18;21). Some of the other possible imbalanced segregants could theoretically be viable, and the reader may wish to determine which ones these would be. This is due to the fact that many of these combinations have a genetically “small” imbalance. All partial trisomies and some partial monosomies for segments of chromosomes 18 and 21 can be viable as a single imbalance; and, when two different imbalances occur in combination, for example, partial trisomy
FIGURE 5–15 Several viable unbalanced forms. The karyotype is illustrated (top) of a mother carrying the translocation t(18;21)(q22.1;q11.2). She had a miscarriage due to adjacent-1 segregation, an abnormal child with a tertiary trisomy, and a stillborn child with a tertiary monosomy, as depicted in the cartoon karyotypes. An uncle with Down syndrome may have had the same adjacent-1 karyotype as in the second row, or possibly interchange trisomy 21, as depicted in the bottom row. (Case of M. D. Pertile.) 92
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
21 plus partial monosomy 18, a pregnancy may still be capable of proceeding substantially along its course. NO UNBALANCED MODE POSSIBLE
Finally, for the translocation in which the quadrivalent is characterized by long translocated and long centric segments, no mode of segregation could produce a viable unbalanced outcome. We emphasize the point that many reciprocal translocations (including whole-arm translocations) are in this category. Consider the family depicted in Figure 5–16, in which a 4;6 translocation t(4;6)(q25;p23) was discovered by chance at amniocentesis. The quadrivalent would have the form depicted in Figure 5–5d. It possesses none of the criteria that would allow a viable imbalance to result, by whatever mode of segregation. The translocated segments are both large (leading to double-segment imbalance); the centric segments are very large; and the content of all four chromosomes is large. Miscarriage is as far as any unbalanced conceptus could ever get. The large kindred of Madan and Kleinhout (1987) graphically illustrates this circumstance: 11 carriers of a t(1;20)
(p36;p11) had had two or more miscarriages, and numerous normal children, but none had an abnormal child. In some such translocations identified fortuitously, for example at amniocentesis for maternal age, there may be little or no history of apparent reproductive difficulty. Meiosis II Nondisjunction. The great majority of segregant forms will have been determined at meiosis I. Meiosis II is not to be completely overlooked, however. A balanced complement may have been transmitted at meiosis I, but a nondisjunction at the following second meiotic division could then produce a gamete with an extra copy, or no copy, of one or other of the derivative chromosomes. In consequence, the conception would have either a partial trisomy of the component parts of the additional derivative chromosome, or a partial monosomy (Masuno et al., 1991). Illustrating the former possibility, Wu et al. (2009) document the case of a father who carried a t(9;15)(q34.3;q13), and whose child, who developed severe autism, had the karyotype 47,XY,t(9;15),+der(15). This imparted a duplication of the proximal long arm of chromosome 15 (which contains autism-susceptibility genes), and
(a)
(b)
FIGURE 5–16 No unbalanced product viable. (a) Pedigree of a kindred in which mother and daughter have had multiple miscarriages, each having (b) the karyotype 46,XX,t(4;6)(q25;p23). (Case of A. J. Watt.) The presumed pachytene configuration during gametogenesis in the heterozygote would be as in Figure 5–5d (chromosome 4 chromatin, open; chromosome 6 chromatin, crosshatched) and, with large centric and translocated segments, the translocation has none of the features that enable viability of any unbalanced segregant combination. Autosomal Reciprocal Translocations •
93
of an 8 Mb segment on distal 9q. This type of “secondary nondisjunction” is very rarely observed.8 Meiotic Drive. As well as the effect of in utero survivability discussed earlier, the nature of the quadrivalent may, of itself, influence segregation. The propensity for a particular segregation outcome may reflect a particular geometry of the quadrivalent, and what sort of ring or chain it forms. Quadrivalents that have translocation chromosomes with short translocated segments more usually form a ring and have the quality of being more likely to generate adjacent-1 gametes, while those with short centric segments, more often existing as a chain, may have a predisposition to the formation of adjacent-2 and 3:1 gametes (Faraut et al., 2000; Benet et al., 2005). This predisposition to form particular classes of segregant gamete may be considered a form of “meiotic drive.” As we have had cause to comment more than once, each translocation is entitled to its individuality and need not necessarily follow the “rules” set out earlier. Faraut et al. (2000) identified a few translocations that “should” have produced sperm with certain expected proportions of adjacent-1 and adjacent-2, but which did not. We have seen a remarkable family in which, over some 10 years of marriage, the woman had innumerable very early miscarriages, about eight at 12–14 weeks, one at 16 weeks, and one phenotypically normal son. The husband (and the son) had the translocation 46,XY,t(12;20)(q15;p13). Perhaps, the quadrivalent was configured in such a way that alternate segregation was very difficult to achieve, and so almost all sperm had an unbalanced complement. De Perdigo et al. (1991) report a possibly similar case, in which they propose that heterosynapsis in the quadrivalent permitted spermatogenesis to proceed, but at the cost of producing many unbalanced gametes. In a family reported in Groen et al. (1998) with a mother having the karyotype 46,XX,t(5q;6q), q35.2;q27), seven sequential retarded siblings of hers are presumed to have had a dup(5)/del(6) karyotype, and only the two eldest and the youngest were phenotypically normal. Observations from the PGD laboratory are further illustrating the point that translocation carriers with very poor reproductive histories may indeed reflect a very high rate of
meiotic malsegregation. The patient in Conn et al. (1999) mentioned earlier, she having the karyotype 46,XX,t(6;21)(q13;q22.3), had had four miscarriages and one child with interchange trisomy 21. She came to PGD, and not one of two oöcytes and nine embryos were chromosomally normal (mostly 3:1, some adjacent segregations). Failure to Form Quadrivalent. Where very small segments are involved, the imperative may lack for the coming together of the four chromosomes with segments in common. This might pertain, for example, to a translocation such as the t(14;15)(q12;q12) in Burke et al. (1996), in which the derivative chromosomes each comprise almost an entire chromosome 14 and chromosome 15, respectively. The 14 and der(15), and the 15 and der(14), might simply synapse as bivalent pairs. The same may very well apply to the general case of the subtelomeric translocation, as discussed earlier. If that were indeed so, then a segregation ratio of 1:1:1:1 would presumably operate, for normal, balanced, and the two imbalanced outcomes: clearly, a high-risk circumstance. Different grounds for the nonformation of a quadrivalent may exist if one chromosome is a very small one. While the three other chromosomes could have come together as a trivalent, the fourth very small one might fail to be captured by the meiotic mechanism. That being so, it could then segregate at random. This could imply a high risk, and might be the reason, for example, that the t(12;13) carrier mother in Figure 5–12 had two out of her four children with a tertiary monosomy. But this is speculative. Detaching of the small derivative from the quadrivalent is an alternative possibility, as discussed in the next paragraph. Parental Origin and Parental Age Effect. There are more women who have been mothers (whether the children are normal or not) than there are men who have been fathers in translocation families. In their review of 1597 children in 1271 translocation families, Faraut et al. (2000) found the mother to be the carrier parent in 61% of the adjacent-1 children, 70% of the adjacent-2 children, and in as many as 92% of the unbalanced offspring from 3:1 segregations. This 3:1 association may reflect an actual
8 Another route to this observation could be a 3:1 disjunction following a crossover in an interstitial segment (Petković et al., 1996).
94
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
maternal predisposition. With advancing maternal age, and after some decades of being held in meiosis I prophase, the small supernumerary chromosome may be increasingly likely to detach from the quadrivalent, and then to migrate at random to one or other daughter cell, when meiosis reactivates in that particular menstrual cycle. On the other hand, no maternal age effect applies to adjacent-1 or adjacent-2 offspring. Here, the maternal excess may more accurately be termed a paternal deficiency, due to reduction in fertility of the male heterozygote (see later discussion). No paternal age effect is discernible in any segregation mode.
microarray in being able to interrogate the whole genome. All 14 prenatally diagnosed de novo translocations in this study were balanced, on this methodology. Baptista et al. (2008) undertook a similar microarray exercise in 31 normal and 14 abnormal cases, making comparison against the absence or presence of genomic imbalance and gene disruption. Some in the abnormal group had microdeletions (although not all were at the breakpoint sites) that offered a likely explanation for the observed phenotype, but none of the normals did. Intriguingly, the frequency of gene disruption was similar in the two groups, implying that a loss of function (presumably) of one copy of some genes may be without phenotypic consequence.
THE PRACTICAL PROBLEM OF THE DE NOVO APPARENTLY BALANCED TRANSLO C ATIO N
I N F ERT I L I TY
A not uncommon problem encountered in the genetic clinic is that of the de novo apparently balanced translocation, which has been discovered in the course of investigation of a child with a nonspecific picture of cognitive compromise and sometimes also some dysmorphic signs. Is the translocation causative, or simply coincidental? Families like those reported in Hussain et al. (2000) offer useful illustration: in this example, an apparently balanced translocation that was cosegregating with a phenotype of nonsyndromic mental retardation. Presumably this translocation, a t(1;17)(p36.3; p11.2), had been de novo at some prior point, possibly with the 65-year-old grandmother of their index case. In this family, there were children and grandchildren, seven of them, to bear witness to the harmful role of the translocation. Thus, the point is underlined: at least some apparently balanced translocations are indeed the cause of the nonspecific clinical picture with which they are associated. In many, however, when these translocations have arisen de novo, it will not be possible, based on classical cytogenetics, to state that the link with phenotypic abnormality was causative, or whether simple coincidence might be the explanation. Newer technologies may now cast light. Using array-CGH with FISH, De Gregori et al. (2007) undertook a systematic search and showed that 40% of 27 apparently balanced de novo translocations in abnormal individuals were, in fact, not so, with microdeletion demonstrable at the sites of breakpoint. In some, they identified de novo deletions at other chromosomal sites than at the translocation breakpoints; and they thus point to the value of
Infrequently, the process of gamete formation in the male translocation heterozygote is disturbed to the extent that gametogenic arrest results. In the analysis of reproductive outcomes in the translocation families of Faraut et al. (2000), looking at prenatal diagnoses in order to avoid bias, 61% of all fetuses came from a carrier mother, versus only 39% from a carrier father; this ratio presumably reflects male infertility associated with the carrier state. This infertility is generally not something that is predictable from the nature of the translocation, and indeed the same translocation may compromise fertility in only some men in the family. Presumably there is, in addition, an effect of the genetic background otherwise (Rumpler, 2001). The detrimental process is considered to be a consequence of failure of pairing (asynapsis or heterosynapsis) of homologous elements in the translocation chromosomes during meiosis I, which promotes association of the quadrivalent with the X-Y bivalent, also known as the sex chromosome vesicle (Paoloni-Giacobino et al., 2000b). The more frequently this association occurs, the more marked the effect upon sperm count. The semen profiles of translocation carriers may not always predict fertility outcomes. In the two cases reported by OliverBonet et al. (2005), one male carried a t(10;14), was normozoöspermic, but had 30% of spermatocytes showing synaptic pairing abnormalities; the other was a t(13;20) man, who was azoöspermic, and showed synaptic pairing abnormalities in 71% of meiotic spreads. This latter carrier also showed decreased recombination frequencies. In men with intact fertility, the spatial organization of Autosomal Reciprocal Translocations •
95
chromosomes within the sperm nucleus differs from normal controls (Wiland et al., 2008). Rearranged chromosomes are not able to be packaged as neatly as they should, so to speak; and this might, in some men, be an additional contributory factor compromising spermatogenesis. The sex difference in susceptibility is striking in the family of Paoloni-Giacobino et al. (2000b). A mother was a t(6;21)(p21.1;p13) heterozygote, and she had eight children, four sons and four daughters (and two miscarriages). The four sons, each one 46,XY,t(6;21), were all married, one three times, and none had any children. Each had severe oligospermia or oligoasthenoteratospermia, and two having testicular biopsies manifested spermatogenic arrest at meiosis I prophase, with extensive asynapsis of several chromosomes. Two sisters were 46,XX,t(6;21), and the one who was married had had two children (and two miscarriages). Oögenesis may not, however, be entirely immune to the translocation obstacle (Speed, 1988; Mittwoch, 1992). Tupler et al. (1994) report two women, one with primary and the other with secondary amenorrhea, who had a balanced reciprocal translocation. Ovarian biopsy in the former, whose translocation was a de novo one, showed absence of the follicle structures in the cortex. Such cases may exemplify a rare translocation effect in the female; equally, given the frequency in the population of the translocation heterozygote, it does remain possible that the link is coincidental rather than causal. Assisted Reproduction. Assisted conception may enable infertile men with a translocation to become fathers. But of course the translocation will, in any event, convey a genetic risk. Meschede et al. (1997) report a man with a t(1;9)(q44;p11.2) having intracytoplasmic sperm injection (ICSI), and two embryos were successfully transferred. At prenatal diagnosis, one twin had an adjacent-1 imbalance conferring a 9p trisomy, the other being 46,XX, and the parents chose selective abortion. Belin et al. (1999) describe a triplet pregnancy achieved via ICSI, the father being a t(20;22) heterozygote. Two normal babies were born (one karyotypically normal, one with the translocation), but the third, with a dup(20p)/del(22q) imbalance, was severely malformed and died in the neonatal period. Meschede et al. (1997) suggest that the population of fertilizing sperm from carriers may differ between ICSI and natural conceptions, and that the risk criteria generally set forth in this chapter should 96
•
be used with caution in couples needing ICSI. Could a chromosomally unbalanced but morphologically normal sperm have a better chance of being selected at ICSI than of succeeding in the open competition that obtains in vivo? We have yet to learn. RA R E CO M PL EXI T I ES
Translocations with Breakpoints at Vital Loci. The great majority of breakpoints in familial translocations between autosomes are apparently sited at points within the genome where they have no effect on its smooth running. Thus, the balanced carriers are phenotypically normal. Rarely, the act of breakage and reunion might compromise a gene or genes; naturally, this is particularly observed in de novo, not familial cases. Compromise could be due to disruption or deletion of genes, or it might reflect a position effect. A detailed example of gene disruption is provided in Kurahashi et al. (1998). A child with lissencephaly (a severe structural brain abnormality) had a t(8;17)(p11.2;p13.3), in this case de novo. The p13.3 breakpoint on the chromosome 17 was sited within intron 1 of the LIS gene, with the gene being split between the two derivative chromosomes: its 5’ part on the der(8), and the rest of it on the der(17). The gene could not, in consequence, function. A similar mechanism is likely the basis of the cognitive and motor delay, and tracheo-esophageal atresia, in a girl with a t(6;15) studied by Giorda et al. (2004), the disrupted gene being the muscle and brain specific gene BPAG1. In the extraordinary coincidence of a recessive mutation on the intact homolog, a translocation breakpoint that disrupted a gene would lead to the appearance of the recessive syndrome, as Kuechler et al. (2010) exemplify in a teenage girl with gonadal failure, who received an apparently balanced t(2;8)(p21;p23.1) from her mother that removed two exons from the FSHR gene (FSH receptor gene, which is located at 2p21), and a point mutation in that same gene on her paternal chromosome 2. As for the position-effect scenario, there are numerous examples, in which a specific phenotype is caused due to a close-by intact gene failing to function. We illustrate in Figure 19–12 (in Chapter 19) one of the earliest such cases, due to a chromosome 17q25.1 translocation whose breakpoint is 50 kb away from the SOX9 locus, leading to campomelic dysplasia (Wagner et al., 1994). More recent cases include a translocation with an 11p13 breakpoint
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
that translocates the PAX6 gene into a chromosomal environment which does not permit its normal expression, with consequential abnormal development of the iris (aniridia); a girl with severe speech impairment who had a t(7;10)(q31;p14) influencing the FOXP2 language-acquisition gene (see later) at 7q31; and a t(12;17)(p13.3;q21.3) affecting the function of the HOXB gene cluster, causing mental retardation and skeletal malformations (Crolla and van Heyningen, 2002; Yue et al., 2007; Kosho et al., 2008). A salutary tale comes from the study of a family with an apparently dominantly inherited syndrome of skeletal anomalies, in which previous cytogenetic tests had given normal results (Stalker et al., 2001). Only after the birth of an infant with severe multiple malformations with an unbalanced karyotype was the fact revealed of a balanced t(13;17)(q22.1;q23.3) cosegregating with the phenotype of the syndrome in the family. There is a fair case for considering that a “bone locus” at 17q23.3 had been disrupted or otherwise influenced by the translocation. Stalker et al. rightly comment that a chromosome test is always worth doing in the investigation of an apparently new familial syndrome, earlier reports of normal cytogenetics notwithstanding, especially if the original laboratory material is not available for review. However, there does always remain the simple possibility that a breakpoint and a disease locus are closely linked, and so the translocation and the disease cosegregate in the family (Hecht and Hecht, 1984). Constitutional translocations might convey a risk for cancer if, for example, a tumor suppressor gene is disabled, or an oncogene is separated from its controlling region. Translocations possibly implying risks for breast cancer, renal cancer, neurofibromatosis type 2, and hematological malignancy are noted in the section on “Genetic Counseling.” Using Translocations to Track Genes. Translocations associated with mendelian disease can serve as a very helpful signpost in mapping the gene. The t(16;22) segregating in a family with polycystic kidney disease mentioned earlier enabled this gene, PKD1, to be isolated: the breakpoint was sited within the gene. Neurofibromatosis type 1 and
Sotos syndrome are other common genetic conditions to have been mapped this way (Ledbetter et al., 1989; Kurotaki et al., 2002). Less readily tractable is a condition such as manic-depressive disease (bipolar disorder), in which an incompletely penetrant mendelian gene is a suspected contributory cause. Baysal et al. (1998) studied a family in which five persons in three generations were affected, each of whom carried (as did some unaffected relatives) a t(9;11)(p24;q23.1). A detailed molecular dissection failed to find disruption in any of a number of plausible candidate genes within the breakpoint regions of 9p or 11q; and so the question remains open whether segregation of the translocation with bipolar disorder was causal or coincidental. When two cases are reported of a mendelian disorder both having a translocation with one breakpoint in common, naturally the case is very much stronger than with a single observation. Thus, there is merit in the proposition of McGhee et al. (2000) that the Coffin-Siris syndrome locus must be at 7q32-q34, they having a patient with a de novo apparently balanced t(7;22)(q32;q11.2), and there being another child on record who had a de novo t(1;7)(q21.3;q34). A formal review of the usefulness of apparently balanced rearrangements in leading the way to locus discovery is given in Bache et al. (2006). The Danish national register of translocations provided a large database, going back to 1968, from which to seek possible disease associations, both mendelian and of complex genetics. Earlier work from this group is outlined in Table 5–3, a listing of breakpoints associated in more than one case of mental retardation; some of these loci may harbor genes important for brain development.9 A similar exercise is proceeding in the United States, focusing more on associations of multiple malformations and mental retardation, and enabling these workers to identify new genes (Higgins et al., 2008). Carrier Couple. Since reciprocal translocation heterozygotes are not uncommon in the population, on rare occasions both members of a couple will, by chance, carry a translocation (Neu et al., 1988b). We have seen, for example, a couple who had had several miscarriages, from 5–9 weeks gestation.
9 The ability to recognize a musical note—“perfect pitch”—is surely due to a brain gene, but this may be an asset, rather than a handicap; 7p22 may be the site of this genetic effect, with three unrelated musically gifted Danish translocation heterozygotes sharing this breakpoint (Bache et al., 2006).
Autosomal Reciprocal Translocations •
97
Table 5–3. Breakpoints in De Novo Rearrangements Associated with Mental Retardation with or without Multiple Congenital Malformations, Observed in More Than One Patient, from a Large Scandinavian Database 1q31 1q44 2q23 4p16 4q22
4q35 5q13 6q27 7q22 7q36
9p24 9q34 12q15 13q34 14q32
Notes: Some of these breakpoints may indicate the sites of disease genes. The 15 breakpoints were seen in 39 patients among 216 with a “disease associated balanced chromosome rearrangement” discovered in 71,739 cytogenetic analyses. Source: Data from Bugge et al. (2000).
The husband’s karyotype was 46,XY,t(7;11) (q22;q23) and the wife’s 46,XX,t(7;22)(p13;q11.2). Presumably, their history of miscarriage reflected at least one parent transmitting, with each pregnancy, an unbalanced gamete: rather many unbalanced karyotypes, as the reader can determine, are possible! A normal child is possible if each contributes a normal or a balanced gamete to the same conceptus. It should, in theory, be reasonably likely in a given conception for the two contemporaneous gametes to have arisen from alternate segregation—as an educated guess, the chance might be about 20%—although at the time of our seeing this family only miscarriage had occurred. A child of theirs having each parental translocation would qualify as having a “complex chromosome rearrangement,” and we shall follow that case in Chapter 12 (and see Fig. 12–6). If a translocation is in a family, and a couple are related, the possibility is open that they might both be carriers. Such a circumstance is illustrated in Kupchik et al. (2005), who report a husband and wife with the karyotypes 46,XY,t(16;18)(p13.3;p11.2) and 46,XX,t(16;18)(p13.3;p11.2). Their child received two copies of the der(18) and one of the der(16), due to alternate segregation in one parent and adjacent-1 in the other. As the reader may determine, the end result was a duplication of distal 16p and a deletion of 18p. In Martinet et al. (2006), a first cousin couple each carried a t(17;20) (q21.1;p11.21), and their severely malformed fetus 98
•
was homozygous for the translocation. The phenotype may have been due to a recessive gene or genes. Similar scenarios with respect to a Robertsonian translocation, and to an inversion, are noted in Chapters 7 and 9, respectively. Mosaicism. Almost all balanced reciprocal translocations are seen in the nonmosaic state. This reflects either that the translocation had been inherited from a carrier parent, or that the rearrangement had arisen preconceptually, in one or other gamete. Rarely, a balanced translocation can be generated as a postzygotic event, and the person is a 46,t/46,N mosaic. In a literature review, Leegte et al. (1998) recorded 29 such cases. One of their subjects, for example, was a man who had presented with infertility, and he had the balanced karyotype 46,XY,t(9;15) (q12;p11.2). His mother had this translocation in a minority of cells on peripheral blood analysis, with the karyotype 46,XX,t(9;15)(q12;p11.2)/46,XX; thus, she was revealed as a somatic-gonadal mosaic. Wang et al. (1998) report a mother mosaic for a whole arm translocation, 46,XX,t(10q;16q)/46,XX, who had a child with a presumed uniparental disomy 16 phenotype from postzygotic “correction” of interchange trisomy 16. The first example of mosaicism for a cryptic translocation is reported in Dupont et al. (2008). Gametic Complementation. Coincidentally abnormal gametes coming from each parent (Fig. 3–6c in Chapter 3) is an extremely rare observation. Park et al. (1998b) describe a unique example of this scenario in the context of a parental translocation. A father with a balanced t(3;15)(p25;q11.2) transmitted a sperm from adjacent-1 segregation, the der(15) being a very small chromosome. This would have led to a near-complete monosomy 15 in the conception. But this was “corrected” by the mother’s transmitting an egg with disomy 15, most probably from a meiosis I error. The child had a partial trisomy for the very small segments 3p25-pter/15pterq11.2 due to the der(15) as a 47th chromosome, but the apparently typical Prader-Willi clinical picture spoke for the predominant contribution to his phenotype deriving from the maternal uniparental disomy 15. Unstable Familial Translocation. Tomkins (1981) documents a family in which a mother with 46,XX,t(11;22)(p11;p12) had one daughter with the same translocation, and another daughter
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
with 46,XX,t(11;15)(p11;p12), and a very few other similar cases are on record. Typically, the translocation breakpoints are at telomeres, centromeres, or in nucleolar organizing regions. There is some sequence similarity in these regions between different chromosomes, and this may set the stage for these very rare “second translocation” events (and see section on “Jumping Translocation,” p. 159).
GENETIC COUNSELING The counselor may have to deal with these questions: 1. Is there a risk of having an abnormal child? 2. If so, what is the magnitude of the risk? 3. What would be the abnormality, and would the child survive? 4. What if the same translocation that I have is found at prenatal diagnosis? 5. What is the risk for pregnancy loss through abortion? Is pregnancy possible? 6. Anything else I should know?
DOES A RISK EXIST OF HAVING AN ABNORMAL CHILD? If a family is ascertained through a liveborn aneuploid child, that very fact demonstrates viability for that particular aneuploid combination. It could happen again. If, on the other hand, the family was ascertained by miscarriage or infertility, or fortuitously, and there is no known family history of an abnormal child, the picture is less clear. Most likely, no aneuploid combination is viable. Alternatively, a viable imbalance may be possible, but it has not yet happened; or an imbalance could occasionally be viable, but usually it is not, and (so far) has led only to abortion. The approach, here, is to determine the potentially unbalanced segregant outcomes, according to the favored mode of segregation—adjacent-1, adjacent-2, or 3:1—and check to see whether any is on record in a pregnancy that produced an abnormal child. Valuable sources of information are Schinzel’s catalog (2001), the European Cytogeneticists Association register of unbalanced chromosome aberrations (ECARUCA), and the U.K. Chromosome Abnormality Database (UKCAD). Where a single-segment imbalance is a potential outcome in a conceptus, from adjacent segregation, and if the potential imbalance comprises an
aneuploidy equal to or less than one of these segments on record, viability must be assumed to be possible. If the potential imbalance comprised an aneuploidy greater than any on record, viability would be unlikely, especially if the aneuploidy is much greater. The great majority of double-segment imbalances from adjacent segregation due to a translocation, ascertained other than by a liveborn aneuploid child, would be expected to lead to lethality in utero. Nearly always, a new double-segment exchange presenting at the clinic will truly be new, and there will be no literature record of exactly the same thing to which the counselor may appeal. Some tertiary trisomies from 3:1 segregation are listed in Schinzel’s catalog, or in ECARUCA or UKCAD, but in many instances one has to make an educated guess, erring on the side of caution, whether the combination of partial trisomies from a derivative chromosome might, in sum, be viable.
The Magnitude of Risk If, in a family, it is judged that there does exist a risk to have an abnormal child, a broad estimate of the level of risk may be derived from a consideration of these factors: the assessed imbalance of potentially viable gametes; the predicted type of segregation leading to potentially viable gametes; the mode of ascertainment of the family; and in 3:1, the sex of the transmitting parent. Most risk figures fall in a range from 0% to 30%; higher risks are rare. These percentages are expressed in terms of abnormal live births as a proportion of all live births, although there are other ways of looking at the risk (see section on “Risk at Time of Prenatal Diagnosis”, and Table 4–1). Overall, the risk is higher in cases ascertained through an abnormal child, versus those identified through other routes; in the review of Youings et al. (2004), the respective pooled figures were 19% and 3%. A precise risk estimate needs to be based on the actual cytogenetic imbalance. Different chromosomal segments contain, of course, different genomic information. It is scarcely possible to come up with a unifying format, given that chromatin is not uniform; as Cohen et al. (1994) comment, “it would be hazardous to suggest a simple mathematical relationship between unbalance length and viability.” Some segments, in the trisomic state, impose a lesser degree of compromise on the process of embryonic development; such as, for example, 18p, and distal 5p. Other segments, Autosomal Reciprocal Translocations •
99
although they may be of shorter length, are lethal during early pregnancy and lead to miscarriage. Some translocations can have their own peculiar segregation characteristics, which a priori were quite unpredictable. Nonetheless, it is interesting to attempt a correlation of quantitative chromatin imbalance with risk to have a liveborn affected child. Daniel et al. (1989), Cans et al. (1993), and Cohen et al. (1994) have compared the haploid autosomal length (HAL) with viability in translocation families. Most (96%) viable imbalances comprise up to 2% monosomy, and up to 4% trisomy, with combinations of monosomy/trisomy viable only when the additive effect of x% monosomy plus y% trisomy falls within a triangular area defined by joining the 2% and 4% points on the x and y axes of a graph (Fig. 5–17). A few (4%) fall outside of this area, and these cases define the boundaries of a “surface of viable unbalances,” reflecting the effects of qualitative differences in different segments of chromatin.
6
% trisomy
5 4 3 2 1 0 0
1 2 3 % monosomy
4
FIGURE 5–17 Viability of combined duplication/ deletion states, according to amount of imbalance, measured as % HAL. Most (96%) fall within the triangular area whose hypotenuse lies between 4% duplication/0% deletion and 2% deletion/0% duplication, and a few outliers define an envelope of viable imbalances. (From O. Cohen et al. 1994, Viability thresholds for partial trisomies and monosomies. A study of 1,159 viable unbalanced reciprocal translocations, Human Genetics 93: 188–194. Courtesy O. Cohen, and with the permission of Springer-Verlag.)
100
•
Only in the case of recurrent rearrangements does the potential exist for direct extrapolation between families. The representatives of this tiny group are the t(11;22)(q23;q11), of worldwide distribution, mentioned numerous times in this chapter, and common enough that its typical unbalanced form has acquired an eponymous nomenclature, Emanuel syndrome (Carter et al., 2009), and of orders of magnitude less frequent, the t(5;11) (p15;p15) associated with Beckwith-Wiedemann syndrome (Slavotinek et al., 1997), the t(4;8) (p16;p23) (Giglio et al., 2002), and the t(4;11) (p16.2;p15.4) (Thomas et al., 2009). For routine practice in the genetic clinic, we suggest starting off with the unvarnished empiric data for individual chromosome segments collected by Stengel-Rutkowski and colleagues, as set out in their invaluable monograph (Stengel-Rutkowski et al., 1988), and discussed in a review and further illustrated in practice (Stene and Stengel-Rutkowski, 1988; Midro et al., 1992), and to which we have already referred several times earlier. The figures set out in Tables 5–4, 5–6, and 5–7, for the three major categories of malsegregation, are summarized from their monograph, and from additional subsequent data. It will generally give a false sense of precision to use decimal points; a rounded figure will suffice. The paucity of information for some chromosomes has necessitated lumping of data for considerable lengths of a chromosome arm; the risk figures derived in this way are, naturally, composites, and indicative rather than definitive. We assume that, in different families with (apparently) the same translocation, the genetic risks will likely be the same, regardless of what may have been the mode of ascertainment. And of course, the principle always applies: if the counselee’s family is large enough, do a segregation analysis to derive a “private” recurrence risk. The figure given for a segment, say, q31/ q34-qter—in other words, a lumped figure applying to a segment extending anywhere from q31-qter to q34-qter—might be given as <0.8%: in other words, a very small risk. (The “less than” sign in the risk data tables is used for estimates in those translocations where no additional aneuploid child has been born apart from probands.) But this figure might have been based mostly on data from families having a q31 breakpoint. A breakpoint at q34 might happen to exclude a dosage-sensitive region of major effect within q33, and thus imbalance for the slightly
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
smaller segment q34-qter might be of considerably greater viability. The risk figure needs to be interpreted intelligently in the light of what is otherwise known from the literature about the segments in question (consult Schinzel, ECARUCA, or UKCAD), and naturally from observation within the same family. The reader consulting and using these figures, imperfect though they may be, will gain a good sense of the practical principles of estimating risk. New data may come to hand. For example, StasiewiczJarockaa et al. (2004) assembled data from 65 new pedigrees involving 16q, to add to the original 35 pedigrees from Stengel-Rutkowski et al. and the new risk calculations are included in Tables 5–4 and 5–5. As expected, the new data continue to be consistent with the notion that the risk for unbalanced offspring increases with decreasing length of the segments. In another study, the methods of StengelRutkowski et al. were applied to a large pedigree segregating a double-segment t(7;13)(q34;q13), together with a sperm karyotype analysis. Midro and colleagues (2006) were able to predict the chance of a miscarriage or stillbirth from carriers in this family to be 13% and 30%, respectively, whereas direct examination of sperm karyotypes indicated 60% abnormal sperm. The high rate of selection against abnormal karyotypes, applying in particular in the latter part of pregnancy with this particular translocation, resulted in a very low presumed risk (0.3%) of the abnormal outcome of a surviving liveborn (and see Table 5–5). Another tool is “HC Forum” provided on the Internet by Cohen et al. (2001), at http://www. hcforum.net, which uses computerized translocation data from their own and others’ material. This can be used as a helpful check on the correctness of the counselor’s pachytene diagram and listing of imbalanced gametes. Some risk estimates from one or other of these sources may need to be treated with reservation,10 and the counselor should always make his or her own judgment based on first principles, and upon an intelligent assessment of the literature relevant to a particular potential imbalance. A very few translocations, or at least some breakpoints, occur with sufficient frequency that specific risk data can be derived. The obvious example is the recurrent t(11;22). Enough 17p13 translocations were on record to enable Pollin et al. (1999) to work out a group risk figure for distal 17p trisomy
or monosomy (the latter producing Miller-Dieker syndrome). The 17p segments ranged in size from 0.12% to 0.38% of HAL, while the segment from the other chromosome was 0.12% to 0.67%, and the combined trisomic/monosomic fractions from 0.24% to 0.9% of HAL. The risks were rather high: 19% for a child with an unbalanced karyotype, rising to 26% if miscarriage and stillbirth were included. Individual circumstances for different types of predisposing translocations are discussed later. The lowest risk for a surviving abnormal child, namely zero, applies in the case of imbalances of large genetic content, in which in utero lethality would be seen as inevitable; and in families interpreted as being in this category, prenatal diagnosis could be seen as unnecessary (Vauhkonen et al., 1985) or could at least be confined to noninvasive ultrasonography. This essentially no-risk circumstance may apply to a considerable, perhaps the great majority of “translocation couples”. The historic data we presently have on record, and to which we appeal in deriving risk figures, were based on classical karyotyping, of differing degrees of precision. Some of the breakpoints given might be arguable, the more so from older reports, and in some (probably only a few), an inaccurate designation might materially misrepresent the risk assessment. In the future, new data will increasingly be generated from microarray analyses, which would, in principle, allow for a very precise comparison with families attending the clinic (Fig. 5–18). Risk at Time of Prenatal Diagnosis. The risk of detecting an abnormality is higher at prenatal diagnosis than it is at the birth of a live baby. This is because there is differential survival throughout pregnancy. Very unbalanced conceptions will abort before the time of prenatal diagnosis. Daniel et al. (1988) derived an overall figure of about 25% for carriers to have an unbalanced fetal karyotype detected at amniocentesis when ascertainment was through a previous aneuploid child, and about 5% when it was through recurrent miscarriage. The amniocentesis figure is at its highest, 35%, in the carrier whose risk otherwise to have an aneuploid live birth lies in the “medium” range (5%–10%) (StengelRutkowski et al., 1988). To give an example from a specific chromosomal segment, Stengel-Rutkowski et al. record a 6% risk for an imbalance in the liveborn from translocations with a proximal 9p breakpoint,
Autosomal Reciprocal Translocations •
101
3.0 2.5 2.0 1.5 1.0 0.5 0.0 −0.5 −1.0 −1.5 −2.0 −2.5 −3.0
Chromosome 15
3.0 2.5 2.0 1.5 1.0 0.5 0.0 −0.5 −1.0 −1.5 −2.0 −2.5 −3.0
Chromosome 20
FIGURE 5–18 Microarray result suggestive of an unbalanced translocation. In each of the figures, probes are ordered on the x-axis according to physical mapping positions for each chromosome. The y axis shows DNA relative amount at the points defined by each oligonucleotide probe: normal diploid amount is at baseline; duplication is above the baseline, and deletion below. Plot areas are shaded pink for gains and blue for losses. In this example, the patient is duplicated (trisomic) for the segment comprising nucleotides 28,197 through 4,174,609 on chromosome 20 (~4.1 Mb), and deleted (monosomic) for the segment comprising nucleotides 96,846,535 through 100,200,997 on distal chromosome 15 (~3.3 Mb). Note that there is also a common deletion variant at proximal 15q. The array nomenclature is arr cgh 15q26.3(96,846,535→100,200,997)×1,20p13(28,197→4,174,609)×3 mat. A microarray analysis only shows gains and losses, and visualization by fluorescence in situ hybridization (FISH) or banding is necessary to determine that this is an unbalanced translocation. Also of note, a microarray analysis on the balanced carrier mother would not have shown the translocation.
versus a 33% risk to detect an imbalance at amniocentesis. In a series of 57 pregnancies in 40 translocation couples, Barišić et al. (1996) determined an overall risk of 16% to discover an unbalanced karyotype at second-trimester amniocentesis, confirming a higher risk (32%) for couples who had previously had an abnormal child, versus a lower figure (12%) where ascertainment had been because of miscarriage. The counselor should be clear about these different types of risk figure being offered (Table 4–1). Ultrasonography can be used as an adjunctive procedure, with normal nuchal translucency in the first trimester and absence of structural anomalies in the second trimester predicting a normal/balanced karyotype (Sepulveda et al., 2001).
102
•
Risks According to Likely Segregation Mode ADJACENT-1 SEGREGATION, SI N G L E SEG M EN T
Specific risk figures for individual single-segment imbalances are set out in Table 5–4. A notable point is the number of risk figures that are very small, less than 1%. This most likely reflects that many imbalances are almost always lethal in utero, and survival through to term is the exception. In fact, we can say that, in order of frequency, there are imbalances which are (1) invariably lethal, (2) almost always lethal, (3) usually lethal, and—the least frequent category—(4) usually survivable. These risk figures are likely to be valid irrespective of the mode of
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Table 5–4. Specific Risk Figures, Based upon Empiric Data, for Having a Liveborn Aneuploid Child, or a Child Stillborn or Dying as a Neonate,a because of SingleSegment Imbalance from 2:2 Adjacent-1 Segregationb TRANSLOCATED SEGMENT THAT WOULD BE IMBALANCEDC
1.
2.
3.
4.
5.
6.
7.
8.
9.
1pter→1p11–34 1p35 1qter→q11–22 q23–32 q42 2pter→p11–12 p13–16 p21–23 2qter→q11–23 q31–32 q33 q33–35 3pter→p13–14 p21 p22–25 3qter→q12–13.2 q21–27 4pter→p11 p14 p15 4qter→q11–13 q21–34 5pter→p11–12 p13 5qter→q13–21 q22–33 q34 6pter→p11–12 p21.2–24 6qter→q11–16 q21–24 q25–26 7pter→p11–13 p15–21 7qter→q11–21 q22–35 8pter→p11–23 p23.1 8qter→q11–13 q21.2–24.2 9pter→p11.2 p13 p22 9qter→q11–13 q21–33
RISK % LIVEBORN
0 ? 0 <1.3 13.6 0 <2.5 5.7 0 <1.7 20.0 22.9 0 <2.3 28.6 0 <1.1 7.7 15.4 28.6 ?0 0.8 3.3 7.0d ? 7.7 25.0 ? 1.3 ?0 20.0 33.3 4.4 19.1 ?0 <0.8 9.1 40e 2.0 11.1 11.8 25 21.2 0 <0.8
S.D.
+ % STILLBORN, NEONATAL DEATHe
+ 5.1 5.2 3.9
+ 15.0 + 14.3 + 6.7
8.9 7.1
+ 11.4 + 13.6
17.1 5.2 4.5 12
+ 38.5 + 7.7 + 7.1
0.8 2.3 2.6
+ 14.1 + 13.1 + 4.0
7.4 7.2
+ 7.7
1.3
+ 11.8
17.9 15.7 3.0 8.6
+ 33.3 + 4.4 + 4.8 + 7.9
3.5 12.6 2.0 6.1 3.7 8.8 4.4
+20 +9.2 +4.2 +2.4 + 8.3
Table 5–4. (continued) TRANSLOCATED SEGMENT THAT WOULD BE IMBALANCEDC
10.
11.
12.
13. 14. 15. 16. 17.
18.
19. 20. 21. 22.
10pter→p11.1 p12–14 10qter→q11–21 q22–23 q24 q25–26 11pter→p11–13 p14 11qter→q13–22 q23 12pter→p11.1 p12 12qter→q11–15 q21–24.3 13qter→q21–32 14qter→q11.1–31 15qter→q11–15 q21–25 16pter→p11.11 16qter→q11–13 q21–23 17pter→p13.3 p11.1 17qter→q11–12 q21–23 18pter→p11.1–11.2 18qter→q11.1–12 q21 q22 19pter→p11–13.2 19qter→q11–12 q13.2–13.3 20pter→p11.1–11.2 20qter→q11.1 21qter→q11.1–22 22qter→q11.1–13
RISK % LIVEBORN
S.D.
4.7 2.6 18.8 9.7 ?0 <1.4 5.9 2.6 14.0 4.9 ?0 <3.1 <2.6 7.0 3.9 9.4 5.2 9.1 8.7 0 <1.5 1.6 1.1 1.0 1.0 0 2.7 2.7 8.3 3.6 6.2 6 <5.4 18.9 3.5 <2.7 ?0 10.0 6.7 ? (probably high) 2.5 2.5 2.9 2.8 15.0 7.8 ?0 ?0 11.1 6.1 20.0 8.0 ?0 13.8 6.4 <2.6
+ % STILLBORN, NEONATAL DEATHe
+ 4.7 + 18.8 + 5.7 + 9.4 + 12.0 + 6.3 + 18.6 + 3.1 + 18.2 + 3.8
+ <3.1 + <5.4 + 7.1
+ 6.7 + 15.0
a Figures are expressed as a percentage of all karyotyped liveborn infants, typically considered as a baby of >28 weeks gestation, with survival at least beyond the neonatal period. Where there are data relating to unkaryotyped stillbirths or neonatal deaths, the figures for these are indicated with a + sign in the third column under “Risk,” as a probable additional component of the overall risk, on the assumption that many, at least, of these cases would have been karyotypically abnormal. The maximum estimate of risk will thus be given by the sum of the two percentage figures. This combined figure may be an overestimate, but if so, likely of small degree; and this may be the more useful figure to consider. b One specific translocated segment is of substantial genetic content (the one shown here), and the other is judged to be of minimal content. For adjacent-1 segregation, the risk does not differ between male and female heterozygotes. For segments not listed here, no specific data are recorded in Stengel-Rutkowski et al. (1988). c Some segments are noted precisely (e.g., 1pter→1p35). Most are given as a pair of breakpoints encompassing a range (e.g., 1pter→1p11–34), extending from a maximum length of terminal-to-proximal breakpoint, to a minimum length of terminal-to-distal breakpoint. Thus, 1pter→1p11–34 refers to an imbalanced segment comprising anywhere from a maximum of 1pter→1p11 (the whole of the short arm) to a minimum of 1pter→1p34 (about one-third of the short arm). d In one reported large family with several cases of “pure” deletion or duplication of this segment (the other segment being derived from acrocentric short arm), the risk was very high: 54% (De Carvalho et al., 2008, and see text). e When the combined live birth + neonatal death figure approximates 50%, this may suggest that the single-segment imbalance is fully viable in utero in either the duplicated or deleted state, with approximately equal numbers of offspring due to alternate and to adjacent-1 segregation. S.D., standard deviation; ?, rare cases have occurred, but data too few to derive a figure;?0, probably no risk; <, no additional aneuploid child has been born apart from the proband, figure is estimate of upper limit of risk interval. Source: From Stengel-Rutkowski et al., 1988, with further entries/amendments from Pollin et al. 1999 (17p13.3), Stasiewicz-Jarocka et al. 2000 and 2004 (1q42, 2q33, 16q), and Panasiuk et al. 2007 and 2009 (4p, 9p), and personal communication from A. Midro 2009 (8p23.1).
ascertainment of the family or of the identity of the other chromosome contributing the telomeric tip, at least in the majority of translocations. By way of example, imagine that a carrier in the t(4;12)(p14;p13) family of Mortimer et al. (1980) noted earlier had sought advice about their own risk to have an abnormal baby. The single-segment involved is 4p14-pter. According to the rules set out earlier, adjacent-1 segregation is the category that implies risk for viable imbalance in this family translocation. Consulting Table 5–4, therefore, we see that the risk for imbalance (whether deletion or duplication) is given as 15.4%. The standard deviation (± 4.5) is quite small, indicating that the estimate is based on a good number of cases. But we also pay attention to the datum “+ 7.7” with reference to unkaryotyped stillbirths and neonatal deaths, many of which will have been, surely,
chromosomally abnormal (probably WolfHirschhorn syndrome). So the true figure to have an abnormal baby at term, who might or might not live, could well be 15.4 + 7.7 = 23.1%. A “private estimate” in this family had come up with a figure of 25%, which is sufficiently close to 23.1% to provide reassurance as to its accuracy. ADJACENT-1 SEGREGATION, DOU B L E SEG M EN T
Every double-segment translocation is likely to be a unique case (or at least no other described family is known), and risk assessment is less precise. One known recurrent double-segment translocation, the t(4;8)(p16;p23), has been seen in sufficient numbers for a useful risk estimate to be derived (Table 5–5). Of course, if the family is large enough,
Table 5–5. Risk Figures for Having a Liveborn Aneuploid Child, or a Child Stillborn or Dying as a Neonate,a because of Double-Segment Imbalance from 2:2 Adjacent-1 Segregation, in 13 Specific Translocationsb RISKC
TRANSLOCATIONS % LIVEBORN
t(1;2)(q42;q33) t(1:3)(q42.3;p25) t(2;13)(p25.1;q32.3) t(3;10)(p26; p12) t(3;15)(q21.3;q26.1) t(4;5)(p15.1;p12) t(4;8)(p16.1;p23.1) t(4;9)(p15.2;p13) t(7;9)(q36.2;p21.2) t(7;13)(q34;q13) t(12;14)(q15;q13) t(16;19)(q13;q13.3) t(16;20)(q11.1;q12)
6.8 63.6 14.5 24.0 ?0 1.6 15 3.2 30 0.3* ?0 1.2 1.1
S.D.
+ % STILLBORN, NEONATAL DEATH
14.5 7.6 8.5
+4.8 +17
3.2 14.5
+ 6.5 +10 + 29.0 +?0
*Plus another 0.2% to account for a theoretical risk for interchange trisomy 13. The considerable gap to the next risk figure, 29%, reflects the several instances in this family of unkaryotyped stillbirths and early neonatal deaths. a Figures are expressed as a percentage of all karyotyped liveborn infants, typically considered as a baby of >28 weeks gestation, with survival at least beyond the neonatal period. Where there are data relating to unkaryotyped stillbirths or neonatal deaths, the figures for these are indicated with a + sign in the third column under “Risk,” as a probable additional component of the overall risk, on the assumption that many, at least, of these cases would have been karyotypically abnormal. The maximum estimate of risk will thus be given by the sum of the two percentage figures. This combined figure may be an overestimate, but if so, likely of small degree; and this may be the more useful figure to consider. ?0 indicates probably no risk, albeit that the 17% risk figure above for stillbirth/neonatal death in the t(3;15)(q21.3;q26.1) indicates viability of the unbalanced state through to the end of pregnancy. b Families published in Kozma et al. (2004), Midro et al. (2000, 2006), Nucaro et al. (2008), Stasiewicz-Jarocka et al. (2000, 2004), Tranebjaerg et al. (1984), and Wiland et al. (2007), and personal communication of A. Midro (2009). c Some figures come from direct segregation analysis, and in others, from applying this rule: halving the risk for the lesser of the two risks, which would otherwise have applied to each translocated segment when viewed as a single-segment imbalance (see text).
Autosomal Reciprocal Translocations •
105
a private segregation analysis will provide the best estimate; and eight other examples are listed in Table 5–5. Otherwise, Stengel-Rutkowski et al. (1988) recommend considering each segment separately. They propose the rule of thumb that the risk will be half that of the smaller of the two risk figures. Even this may be an overestimate; in many cases, the duplication/deficiency from a double-segment imbalance will be invariably lethal in utero—a risk of 0%—notwithstanding that each segment separately is on record with viability in the single-segment state. In one instance, however, this rule was vindicated, in the t(4;9)(p15.2;p13) family listed in Table 5–5. The risk attaching to 9p13, as a singlesegment, is 6.4% (from Table 5–4), and this halved to 3.2%. From an actual family study, the comparable figure was also 3.2% (although a wide standard deviation of ± 3.2) (Midro et al., 2000). Preimplantation genetic diagnosis (PGD) requires a different viewpoint, since in utero lethality has not had the chance to operate, and the denominator of the risk figure is quite different: this now refers to the rate of abnormalities in the day 3 embryo. In a doublesegment translocation t(3;11)(q27.3;q24.3) carried by a brother and sister reported in Coonen et al. (2000), at least 15 out of 18 embryos of the brother were karyotypically unbalanced, and only one was normal or balanced. This one embryo was transferred, amniocentesis showed 46,XX,t(3;11), and a healthy carrier daughter was in due course born. His sister, a carrier of the same translocation, underwent two treatment cycles, with two out of six embryos apparently normal, but neither transferred successfully. ADJACENT-1 SEGREGATION WITH S UBT ELO MER IC D O UBLE-SEGM E N T E XCHA N G E
The viable outcomes will be due to one or possibly both of the dup/del and del/dup combinations, and the family history may well be informative, as illustrated by the t(1q;3p) family reported in Kozma et al. (2004). Either of the combinations could arise from adjacent-1 segregation, or from independent 1:1 segregation of each normal homolog and its derivative chromosome (see Fig. 5–8). From random 1:1 segregation, if that occurred, a theoretical risk of 50% for unbalanced karyotypes would apply at conception. More such cases will require study before a clearer picture can be drawn; but in any event a high risk, and as judged from the family history, is very probable. 106
•
In one PGD case reported, relating to a couple one of whom carried a t(2;17)(subqter;subqter), 13 of 18 embryos showed 2:2 segregation for the translocation (six alternate, seven adjacent-1), consistent with either two independent 1:1 events, or 2:2 disjunction from a quadrivalent (McKenzie et al., 2003). But the fact that the remaining five malsegregants displayed 3:1 disjunction suggests that a quadrivalent may indeed have formed, even if, considering the nature of this translocation, 3:1 is contrary to the “rules” of malsegregation set out earlier. ADJACENT-2 SEGREGATION
Very few translocations are capable of producing viable adjacent-2 segregant products, and the data on specific risk levels are limited (Table 5–6). Where the potential imbalance has considerable viability, for example, trisomy 9p and trisomy 21q, the risk is likely to be substantial and may be in the range of 20%–30%. The carrier mother in Figure 5–9 would
Table 5–6. Specific Risk Figures for Liveborn Aneuploid Child due to Imbalance from 2:2 Adjacent-2 Segregation CENTRIC SEGMENT THAT WOULD BE IMBALANCED
RISK
4. 8. 9. 10. 12. 13. 14. 15. 20. 21.
% ?0 ? 18.4 ? ? ? ? 11.8 27.3 ?
4pter→q11–13 8pter→q12–13 9pter→q11–13 10pter→q11–21 12pter→q11–13 13pter→q14–21 14pter→q21–22 15pter→q13–24 20pter→q11.1 21pter→q11.1–22
S.D.
4.5
7.8 13.4
Note: Figures are expressed as a percentage of all live births. No obvious difference exists according to sex of parent. For segments not listed, no specific data are recorded in Stengel-Rutkowski et al. ?, rare cases have occurred, but data too few to derive a figure; ?0, probably no risk. Source: From Stengel-Rutkowski et al. (1988).
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
have, from Table 5–4, an 18% risk for the recurrence of trisomy 9p. 3:1 SEG R EG ATIO N , T E RT I A RY ANE UPLO IDY
In contrast to 2:2 segregation, the probabilities for unbalanced 3:1 outcomes differ between the sexes, with the female having the greater risk. For translocations other than the common t(11;22)(q23;q11), the risk is generally small and is less than 2%. Nevertheless, each translocation is entitled to its individuality, and atypically higher risks are possible, as may be exemplified in the t(12;13) noted earlier and shown in Figure 5–12, in which two out of four children had a tertiary monosomy. In this case, it could be that the tiny derivative segregated independently, at random. The common t(11;22) 3:1 segregation with tertiary trisomy is practically the only segregation mode to produce a viable abnormal baby in the common t(11;22)(q23;q11) (Fig. 5–11). Different figures have been proposed for the level of risk. From the data of Stengel-Rutkowski et al. (1988), as listed in Table 5–6, the risk is 3.7% and <0.7%, respectively, for the female and male carrier. In a very large collaboration, with data from 110 families seen in 15 countries (there being some overlap with the material in Stengel-Rutkowski et al.), Iselius et al. (1983) arrived at risk figures for the female and male heterozygote, respectively, of 2.1% and 1.8%. Notably, in most of these families the index case was the only one known definitely to have the unbalanced karyotype. However, it could be supposed that reported malformed stillborn infants in these families were rather likely also to have had the unbalanced karyotype, and if this assumption is accepted, the risk figures for a live- or stillborn affected infant would increase to 5.7% and 5%, respectively. A rather higher risk figure for the female carrier, namely, ~10%, is due to Zackai and Emanuel (1980). These authors also observed that the chance of transmitting the translocation in balanced state is significantly greater than the theoretical 50%, with a risk of >70% in the families studied. As a further untoward consequence of being a carrier of this common t(11;22), the female may be at risk for breast cancer ( Jobanputra et al., 2005; Wieland et al., 2006).
3:1 SEG R EG AT I O N , I N T ER CH A N G E A N EUPLO I DY
The risk to have a child with Patau, Edwards, or Down syndrome from an interchange trisomy is remarkably small. It may be in the vicinity of 0.5% in the female, and less than this in the male (StengelRutkowski et al., 1988). Upper limits of the estimated risks are given in Table 5–7. The figures for PGD can be much higher, as shown in Table 5–2, and as illustrated by the case of Conn et al. (1999) noted earlier, in which a woman with the karyotype 46,XX,t(6;21)(q13;q22.3) had 9/9 embryos with chromosome imbalance, including two with interchange trisomy 21, and one with probable interchange monosomy 6. MORE THAN ONE UNBALANCED SEGR EG A N T TYPE
It is probably prudent to assume that where more than one mode of segregation can lead to a viable outcome, the overall risk will be cumulative and will be given by the sum of the individual risks. Thus, the carrier mother of the t(11;18)(p15;q11) shown in Figure 5–14 would have a risk comprising three components: duplication 18q11-qter due to adjacent-1; tertiary trisomy 18pter-q11 due to 3:1; and trisomy 18 due to 3:1 interchange. From Tables 5–4 and 5–7, and choosing the closest listed segments, these risks are 2.5%, <1.3%, and <0.2, respectively, for a total of up to 4.0%.10 I M PR I N TA B L E CH R O M OS O M ES A N D U NI PA R EN TA L D I S O MY
Any translocation, of which a participating chromosome has an imprintable segment, is to be considered from this specific perspective. Engel and Antonarakis (2002) list 10 familial translocations from the literature associated with the birth of a child with a UPD syndrome, including Prader-Willi and Angelman syndromes (PWS and AS), BeckwithWiedemann syndrome, and UPD14; and SilverRussell syndrome due to UPD7 has been reported in association with a maternal t(7;16) (Dupont et al., 2002). The gender of the transmitting parent becomes of relevance: for example, in a family
10 Intuitively, this figure may seem too low, given the well-known viability of partial and complete trisomy 18. The figure of 15% from HC Forum would seem to be more plausible.
Autosomal Reciprocal Translocations •
107
Table 5–7. Specific Risk Figures for Liveborn Aneuploid Child due to Imbalance from 3:1 Single-Segment Segregation A. Tertiary Trisomy or Monosomy SEGMENT THAT WOULD BE IMBALANCED
4. 8. 9.
4pter→q12–13 8pter→q12–13 9pter→q11–32
10. 11.
10pter→q11.1–21 11qter→q23*
12. 13.
12pter→q11–13 13pter→q12–33
14.
14pter→q11.1–24
15. 16.
15pter→q11.1–24 16pter→p11.1
18.
16qter→q11.1-p11.1 18pter→p11.1–21
20.
20pter→q11.1
21. 22.
21pter→q11.1–22 22pter→q11.1–13
RISK
% ? ? 1.7 (mat) ?0 (pat) ? 3.7% (mat) <0.7% (pat) ? 2.6 (mat) 0 (pat) 2.6 (mat) <0.8 (pat) <0.9 <1.8 (mat) 0 (pat) ? (mat) <1.3 (mat) 0 (pat) <4.4 (mat) ?0 (pat) 6.9 (mat) <3.5 (mat) ? (pat)
S.D. 1.7
1.8 2.6
4.7
Phenotype and Survivability
B. Interchange Trisomy CHROMOSOME THAT WOULD BE TRISOMIC
RISK
% <0.2 (mat) 0 (pat) <0.2 (mat) <0.3 (pat) 0.5 (mat) <0.6 (pat)
13 18 21
Notes. Figures are expressed as a percentage of all live births. Risks for maternal transmission (mat) are typically greater than for paternal (pat) in 3:1 segregations. For segments not listed, no specific data recorded in Stengel-Rutkowski et al. ?, rare cases have occurred, but data too few to derive a figure; ?0, probably no risk. *The common t(11;22)(q23;q11); see p. 107. Source: From Stengel-Rutkowski et al. (1988).
108
•
segregating a rcp(4;11)(q35;p15.5), the unbalanced state 46,der(4) was expressed differently in children inheriting the der(4) from father or mother (Bliek et al., 2009). Overgrowth was part of the phenotype with paternal transmission, and growth restriction with inheritance from the mother, reflecting the different imprinted status of the 11p15.5 translocated segment on the der(4). The most notable phenotypic distinction is seen in differential parental transmission of 15q13 imbalances, leading to either PWS or AS. A potential risk for UPD following postzygotic “correction” was noted earlier. What looks like alternate segregation in the fetus could actually have been 3:1 interchange trisomy, with a postconceptual loss of the homolog in question. In practice, this appears to be an exceedingly rare outcome (Dupont et al., 2002; Kotzot, 2008a; Heidemann et al., 2010). An example is the case in Calounova et al. (2006): a child with PWS had the same 46,XX,t(8;15) (q24.1;q21.2) karyotype as her mother, with absence of a paternal chromosome 15 and thus with UPD15mat. Another mechanism, rare indeed, is gametic complementation (see later discussion). This much is certain: any translocation involving chromosome 15 in particular is to be approached very circumspectly. The translocation with a 15q11–13 breakpoint is a special case, and unequal meiotic crossing-over can give a recombinant chromosome cytogenetically indistinguishable from the balanced state (Horsthemke et al., 1996).
S.D.
0.5
A major degree of dysmorphogenesis, involving several body systems, and globally disordered brain function is the typical picture in viable autosomal imbalance. Many patients will come with the knowledge of the particular phenotype of at least one of the viable segregant outcomes—the proband in their own family. The same imbalance in a future pregnancy would be expected to lead to a similar physical and mental phenotype.11 Survivability is less predictable because, for many conditions, there
11 Similar may only mean “somewhat the same.” Just as trisomy 21 presents a considerable range in intellectual capacity, variation may be observed with the identical segment, duplicated or deleted, in different family members. The rest of the (balanced) genome, which will of course differ, may dictate a relative vulnerability, or resistance, to the damaging effects of the imbalance.
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
is a fine line between relative robustness and a fragile hold on existence, intrapartum and postnatally. Whether there is a heart defect (a frequent malformation in many chromosomal disorders) may be a major factor in this. As for the phenotype of potentially survivable outcomes other than those already exemplified in the family, reference to the chromosomal catalogs and databases (de Grouchy and Turleau; Schinzel; ECARUCA; UKCAD) and to the journal literature provides a guide. For imprintable chromosomes, there may be an influence of the parental origin of the aneuploid segment, as noted earlier. THE PARENTAL BALANCED TRANSLO C ATIO N IN T HE F E T U S
The conventional wisdom is that if the same (balanced) karyotype found in the carrier parent is detected at prenatal diagnosis, there is no increased risk for phenotypic abnormality in the child: like parent, like child. Some have doubted this, and Fryns et al. (1992a) measured a 6.4% risk of mental and/or physical defects in the heterozygous children of translocation carriers (this figure including the background risk of 2%–3%). Others remain sceptical and impute ascertainment bias as the confounding factor (Steinbach, 1986). Theoretical mechanisms whereby an apparently balanced translocation could have a deleterious consequence, the parental normality notwithstanding, include the following four: a cryptic unbalanced defect beyond the resolution of routine cytogenetics (but nowadays possibly detectable on microarray); the postzygotic loss of a derivative chromosome in one cell line, converting an unbalanced to a mosaic balanced/ unbalanced state; a position effect; and uniparental disomy. Concerning the cryptic unbalanced defect, Wagstaff and Hemann (1995) provide a disconcerting example: an apparently balanced parental reciprocal translocation which turned out to be a complex chromosome rearrangement, with a tiny segment from the breakpoint of one of the translocation chromosomes inserted into a third chromosome (p. 216 and Fig. 12–7 in Chapter 12). In families in which the balanced translocation has been transmitted to numerous phenotypically normal individuals, such a scenario is most unlikely, since consistent co-segregation of the “cryptic chromosome” to give an overall balanced complement in all these persons would be improbable. Where the translocation is of
more recent origin, perhaps de novo in the parent, the possibility may be more real. As mentioned earlier, the increasing sophistication of molecular cytogenetics may enable such cases to be teased out. Clearly, if this sort of investigation is to be done, it should be well ahead of any pregnancy. The case reported by Dufke et al. (2001) illustrates the possible scenario of mosaicism. An abnormal child with the same balanced t(17;22) (q24.2;q11.23) as his mother on peripheral blood analysis, showed, on skin fibroblast culture, a 47,t(17;22),+der(22) karyotype. This mosaic picture may reflect there having been a “interchange tertiary trisomy”7 complement in the conceptus, with postzygotic loss of one of the two der(22)s in blood-forming tissue. A similar scenario is documented in Prontera et al. (2006): a mother carrying a t(1;15)(q10;p11) had an abnormal child, in whom the same apparently balanced karyotype had been shown at prenatal diagnosis. In view of the abnormal phenotype, a stringent postnatal analysis was done, which revealed a small fraction of cells, 4% (on blood), with trisomy 15; the conclusion is thus drawn that the initial conception had been from a 3:1 malsegregation with interchange trisomy, and a mitotic “correction” thereafter resulted in loss of the additional chromosome 15 in a substantial fraction of cells, but obviously not all. These reports raise the question: could the excess noted by Fryns et al. in a postnatal population be accounted for, in part at least, by this process? And, if so, could this be the basis of a misleading prenatal diagnosis? In fact, it could be imagined that, if the mother in Dufke et al. had had an amniocentesis, the unbalanced 47,t(17;22),+der(22) state would have been seen, since the sampling of amniocytes is somewhat equivalent to taking several skin biopsies. On that premise, it could be argued that a good number of normal cells/colonies from amniocentesis might indicate an unlikelihood of any such mosaicism, albeit that the very low degree of mosaicism in the other case described earlier would argue against complete reassurance in this respect. The concept of “position effect” was discussed earlier. A particular gene in the close vicinity of a translocation breakpoint may function normally in a parent; but, for whatever reason, in the child the gene in question may be silenced, due to an effect of the adjacent chromatin of the other participating chromosome. Realistically, however, a question of position effect is worth raising only in families where the abnormal phenotype has already occurred. Autosomal Reciprocal Translocations •
109
Of similar extreme rarity, there is the theoretical question of uniparental disomy following postzygotic “correction” of an interchange trisomy for an imprintable chromosome. If an important additional risk due to one or other of the aforementioned scenarios really does exist, it is surely very small, perhaps no more than “a fraction of a percent” that a child with the “balanced” parental karyotype might have a defect of mostly unpredictable severity and extent. In the meantime, it remains true that in the great majority the balanced translocation really is balanced, structurally and functionally, and will have, of itself, no detrimental effect. Thus, in practical terms, it would be appropriate to advise continuing a pregnancy when the fetal karyotype is the same as that of the carrier parent, and with very considerable (if not absolute) confidence of a normal outcome.
Infertility and Pregnancy Loss I NFE RTILITY
Occasionally, some male translocation carriers are infertile with a spermatogenic arrest, as discussed earlier. There is little (if any) increased incidence of infertility in the female; from this point of view, oögenesis is a more robust process. MI SCA RRIAG E
Conceptions with large imbalances will abort. Against the background population risk of 15% for a recognized pregnancy to miscarry,12 the risk for the translocation carrier is rather greater and is in the range of 20%–30% (Stengel-Rutkowski et al., 1988). For a few, the risk is very high, well over 50%. An increasing viability of conceptuses implies a correspondingly diminishing likelihood of pregnancy loss by miscarriage. Not to diminish the distress felt at the loss of a welcomed and wanted pregnancy, patients can be heartened that miscarriage, in this setting, is the natural elimination of a severe abnormality, which provides the opportunity to make a fresh, and hopefully, a more fortunate start. For a couple having lost all pregnancies to miscarriage, karyotyping in the previous generation may be helpful. The consultand would, in him or her self,
embody the proof that the heterozygote can have a normal child, should one of his or her parents also be a carrier. Optimism has to be muted, however, in the setting of a family history of many miscarriages, which may indicate a propensity for the production of unbalanced gametes. Preimplantation Genetic Diagnosis. PGD has obvious attraction as a means to avoid a pregnancy with an imbalance, by choosing only embryos with a balanced complement, following embryo biopsy. Indeed, the practical observation is that this approach may substantially improve the chance for a translocation carrier to have a normal child, especially in those who have had several miscarriages and no liveborn child (Otani et al., 2006; and see Chapter 26). As we have commented a number of times previously, the risk figures in this context will be rather different. A chromosomally abnormal embryo at day 3 (when PGD is applied) has not been subject to selection pressure, and so a wide range of imbalances may be seen, the very great majority of which could never survive to term, and many of which would fail even before implantation (day 5). The data in Table 5–2 might suggest a 43% chance for a normal/balanced embryo from a male heterozygote, and 50% from a female. But the outlook may be less promising than this, as more data come to hand; and the average chance to obtain a normal/balanced embryo may be as low as 20% (Goosens et al., 2009). Obviously, couples in this situation will hope that their in vitro fertilization (IVF) team can produce a good number of embryos.
Other Issues Other Family Members with the Same Translocation. It appears to be the case that a translocation studied in one family member will typically display similar meiotic behavior in other carriers in the family; at least, this applies to the male, in whom gametic analysis is more readily pursued (Benet et al., 2005; Wiland et al., 2007). Thus, genetic advice can be, in practice, the same, for one and all. Associated Mendelian Condition. Rare translocations are associated with a mendelian disorder due either to the breakpoint disrupting or influencing a
12 This figure applies respect to clinically diagnosable miscarriage, mostly occurring in the period 8 to 16 weeks gestation. Severely imbalanced forms may be lost as very early, even occult, abortions (p. 382).
110
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
locus, or with coincidental linkage to a mutation near the breakpoint. We note some examples in the earlier section on “Biology.” In such families, over and above any risk associated with unbalanced segregants, one should discuss the risk of transmitting the abnormality peculiar to that chromosome. Cancer Risk. In rare familial translocations, the rearrangement may promote mitotic malsegregation, or disrupt a tumor suppressor gene, and thus comprise a “hit” in the cascade of events leading to the cellular phenotype of cancer. A well-recognized case is that of chromosome 3 translocations implicated in familial renal cancer, of which a number of examples have been published (Valle et al., 2005). According to one construction, a three-hit sequence is envisaged, the first hit being the actual inheritance of the balanced translocation. Then, the mechanism is a mitotic malsegregation in an embryonal kidney cell. The derivative chromosome containing the 3p segment is lost, and in consequence one daughter cell, and thus the lineage from it, has only one copy of distal 3p, on the normal homolog (the second hit). Thereafter, on this remaining normal chromosome, a somatic mutation occurs in postnatal life at a 3p-located tumor suppressor gene in a kidney cell within this lineage (the third hit); and now the stage is set for a renal cancer to come into being. Otherwise, a t(3;6) is recorded which possibly predisposes to hematological malignancy (Markkanen et al., 1987). Two families are known with familial adenomatous polyposis being due to a constitutional reciprocal translocation, t(5;10)(q22;q25) and t(5;8)(q22;p23.1), the relevant tumor suppressor gene (APC) having been disrupted at the 5q22 breakpoint (van der Luijt et al., 1995; Koorey et al., 2000). A father and daughter with an apparently balanced t(1;22)(p36.1;q12) both had neurofibromatosis type 2, due to the NF2 gene having been disrupted (Tsilchorozidou et al., 2004). Laureys et al. (1990) reported a child with a de novo t(1;17) (p36.2;q12) presenting with neuroblastoma; it may be that somatic loss of the der(17) and duplication of the normal chromosome 17 in cells of the nervous system set the stage for the tumor to arise (Van Roy et al., 2002). An excess of constitutional rearrangements in a series of children with various tumors suggests the possibility of a causative role for some of them (Betts et al., 2001). The possible increase in risk for breast cancer in the common t(11q;22q) has been noted previously. Where the cytogenetic-cancer associations are firm,
heterozygotes should receive appropriate counseling, and entry into a cancer surveillance program is appropriate. Often, the associations appear to be no more than fortuitous (given that rearrangements are not uncommon, and cancer is very common). Interchromosomal Effect. There had originally been concern that a reciprocal translocation heterozygote might be prone to produce gametes aneuploid for a chromosome not involved in the translocation, specifically, in this context, chromosomes 13, 18, 21, or X, and there has been the occasional report of a translocation carrier having offspring with chromosomal imbalance not related to the family’s translocation (Couzin et al., 1987). Warburton (1985) reviewed the associations of reciprocal translocations and trisomy 21 from unbiased (amniocentesis) data and found no evidence to support the contention. Uchida and Freeman (1986) and Schinzel et al. (1992) studied families in which a child with trisomy 21 also had a balanced translocation, and while in several the translocation was of paternal origin, in fact the extra chromosome 21 came from the mother. More directly, numerous sperm karyotyping studies have, for the most part, shown no increase in disomies unrelated to the translocation, although some workers have raised doubts (Estop et al., 2000; Oliver-Bonet et al., 2004; Machev et al., 2005). Pellestor et al. (2001) suggest that carrier males with poor semen indices are the only ones in whom any such effect might exist; in which case, it might be the altered testicular environment, rather than the translocation of itself, that is the cause (Kirkpatrick et al., 2008). Analysis of embryos at preimplantation diagnosis supports the case against any effect (Gianaroli et al., 2002). Possibly, some specific translocations may have a very small individual risk, but there seems little reason to withdraw from the generality of Jacobs’ assessment from 1979, and in practical terms we expect this view to prevail: “there is no indication that parents with a structural abnormality are at an increased risk of producing a child with a chromosomal abnormality independent of the parental rearrangement . . . [and] their recurrence risk for such an event is the same as the incidence rate in the population.” Only with infertile men (needing ICSI for conception) might there really be a small risk, and as noted earlier, this may be due more to the infertility per se. No doubt the debate will continue.
Autosomal Reciprocal Translocations •
111
6 sex chromosome translocations
THE SEX CHROMOSOMES (gonosomes) are different, and sex chromosome translocations need to be considered separately from translocations between autosomes. A sex chromosome can engage in translocation with an autosome, with the other sex chromosome, or even with its homolog. The unique qualities of the sex chromosomes have unique implications in terms of the genetic functioning of gonosome-autosome translocations. Unlike any other chromosome, the X chromosome is capable of undergoing “transcriptional silencing” or, as more usually spoken, facultative inactivation, of almost its entire genetic content. This fact has crucial consequences for those who carry an X-autosome translocation, in both the balanced and the unbalanced states. And unlike any other chromosome, the Y is composed of chromatin which is, in large part, permanently inert. Some translocations of this inert material can thus be of no clinical significance.
112
•
BIOLOGY THE X-AUTOSOME TRANSLOCATION Both females and males can carry, as heterozygotes or hemizygotes, an X-autosome translocation, in balanced or unbalanced state. But the implications for the two sexes are rather different, and we therefore need to treat the two cases separately. First, we need to review the concept of X-inactivation. X-INACTIVATION
The normal female has two X chromosomes, and yet the possession of only a single X is sufficient to produce normality in the 46,XY male. Are the sexes really so genetically different? Does the female really need a second X? The answer is a qualified no.
The second X is largely surplus to requirement, and it is subject to transcriptional silencing. Very early in embryonic existence, around the period of the morula and blastocyst stages, a process is initiated whereby one of the X chromosomes in every cell of the female conceptus is randomly genetically inactivated (van den Berg et al., 2009). This process is called (after Dr. Mary Lyon) lyonization. In all descendant progeny cells thereafter, the same X chromosomes remain inactive or active, respectively. This “dosage compensation” allows for a functional monosomy of most of the X chromosome. Transcriptional silencing is initiated at an X-inactivation center (XIC) in Xq13 (Fig. 6–1), and it spreads in both directions along the chromosome. Within the XIC is a gene XIST that is cis-acting (that is, it can influence only the chromosome that it is actually on), and that is transcribed only from the inactivated X. This transcript, named “XIST,” for X (inactive) specific transcript, is not translated into protein but functions as an RNA molecule. The XIST RNA “coats” the X chromatin and may act first by influencing the degree of acetylation and other
modification of the histones, and this then preventing the DNA from being transcribed. This inactive state is then “locked in” by methylation of CpG islands, and this methylation status remains in place in the descendant daughter cells. The reader wishing full detail is referred to Migeon’s Females Are Mosaics (2007). Normal women can have quite skewed ratios of active Xm and inactive Xp chromosomes1 and vice versa, even more than 90:10, and there can be differences in ratios between different tissues in a woman (Sharp et al., 2000). The inactive X replicates late during the cell cycle; the active X replicates early, along with the autosomes. But this is not to say that the female’s second X chromosome is unnecessary (a rather obvious statement, considering the difference between 46,XX and 45,X individuals). Not all genes on the X chromosome are inactivated, and thus some loci are, in the normal female, functionally disomic. There is a block to the spread of inactivation into the primary pseudoautosomal region (PAR1), which comprises the terminal 2.6 megabases of Xp in band p22.3; this segment has an homologous region on distal Yp
FIGURE 6–1 Notable regions of the sex chromosomes. AZF, azoöspermia factor regions a–d. Dots show specific loci: DAZ, deleted in azoöspermia locus; MLS, microöphthalmia with linear skin lesion gene; SHOX, short stature homeobox gene (X chromosome), SHOXY, short stature homeobox gene (Y chromosome); SRY, testis-determining locus. CR1, 2 show critical regions 1 and 2 (p. 116).
1 Denoting the one from her mother as Xm, and the one from her father as Xp.
Sex Chromosome Translocations •
113
(Fig. 6–1). There is a secondary PAR (PAR2), which extends over 320 kb within distal Xq, having homology with distal Yq (Kvaløy et al., 1994). An obligate recombination event occurs in the PAR1 of the X and the Y chromosome at male meiosis; recombination between the secondary PARs, if it occurs at all, is infrequent. Certain other loci elsewhere on the X than in the PARs (some of which have homologs on the Y) are not subject to inactivation, and disomic expression of these genes in the female (and, for some, in the male) is normal (Disteche, 1995). One such is the nonpseudoautosomal X-Y homologous region, at Xq21.3 and Yp11.2, respectively, and in fact this is the largest region, some 4 Mb in length, of shared sequence between the sex chromosomes (Wilson et al., 2007).
The Female X-Autosomal Heterozygote The balanced X-autosome translocation carrier has two translocation chromosomes, the der(X) and
the der(autosome). The X segment in one of these, most commonly the der(X), contains the XIC, and the X segment in the other, usually on the der(autosome), lacks the XIC. The latter segment, having no XIC of its own and being beyond the influence of the XIC on the other derivative, is always active. The only way, then, for the karyotypically balanced female X-autosome heterozygote to achieve a functionally balanced genome is to use, as her active X complement, the two parts of the X in the two translocation chromosomes: together, they add up to an equivalent whole, and functioning, X chromosome. The other chromosome, the normal X, is inactive. The cartoon karyotype in the 46,X, t(X;12) carrier mother in Figure 6–2 shows the normal X as inactive (dotted outline), and the X-segments of the der(X) and der(12) as active (solid outline). Probably, the mechanism to bring about this asymmetric inactivation is as follows. Inactivation is initiated at random in each cell, at either one of the XICs. Some cells will be functionally balanced, with the intact X inactive, as described earlier. Others, in
FIGURE 6–2 Inactivation patterns. Mother with a balanced X-autosome translocation, showing patterns of inactivation in herself and in her two chromosomally unbalanced children with partial Turner and partial Klinefelter syndrome, respectively. Dashed outline indicates inactivated chromosome. The inactivation pattern of a theoretical third child with a partial X trisomy is shown at right. Note that the balanced carrier inactivates her normal X chromosome, while it is the abnormal X which is inactivated in the unbalanced offspring (and, in the third child, one of the additional normal X chromosomes as well). Based on family in Figure 6–4. 114
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
which the intact X is active, will have a functional disomy for the X chromosome segment that is translocated to the der(autosome), due to this X segment not being subject to transcriptional silencing, and thus genetically active. According to this theory, cell selection then eliminates the functionally partially disomic X lines (Fig. 6–3, sequence a→b→c). This mechanism is successful in a fraction of translocation heterozygotes, and aside from a possible gonadal effect (see later), such individuals are phenotypically normal. This mechanism, as it would seem, may not infrequently fail, and phenotypic abnormality is the consequence. “Not infrequently” may translate to as much as 25%, with reference to the literature study of Schmidt and Du Sart (1992). If some functionally disomic cells survive and come to comprise part of the soma (Fig. 6–3, sequence a→b→d), this would, presumably, have some deleterious effect. The natural prediction is that only cells with small partial disomies would be capable of survival. Thus, we might more commonly expect to observe, in these affected carrier females, translocation breakpoints
in distal Xp or distal Xq (Xp22 and Xq28), which would impart disomy for only a very small segment of either distal X short arm or distal X long arm. This was indeed the observation in the reviews of Schmidt and Du Sart (1992) and Du Sart et al. (1992). To the contrary, however, the X breakpoints in the data assembled from a survey conducted by the U. K. Association of Clinical Cytogeneticists showed a wider distribution, and the X and der(X) did not necessarily display a skewed inactivation (Waters et al., 2001; A. J. M. Crocker, personal communication, 2001). There was no particular predilection for site of breakpoint, other than Xq22 being overrepresented. These authors believe that their data may be more representative, having avoided a reporting bias. Given their observations, they propose that gene disruption may be similarly important as a factor causing phenotypic abnormality. Part of the difference between the two studies may lie in the nature of the presenting phenotypes, whether these be malformation/cognitive compromise, or merely compromise of ovarian function. More such data would be useful. (c)
(a)
(b) Inactivation
tion
Selec
(d) No s
elect
ion
FIGURE 6–3 Skewing or nonskewing of X chromosome inactivation, as a theoretical explanation for the X-autosome carrier being of either normal or of abnormal phenotype (and see text). (a) Before X-inactivation occurs, both the normal and the der(X) are active in all cells (shown in light gray). (b) X-inactivation occurs as a random, cell-autonomous process. Cells shown in white have the der(X) as the active chromosome, and thus the genetic activity of these cells is balanced with respect to X chromosomal output. The cells shown in dark gray have the normal X-active, and in consequence their X chromosomal activity is imbalanced, due to the additional output from the X-segment of the der(autosome). Subsequently in embryonic development: Either (c) the cells with the normal X-active (dark gray) die out, due to their functional genetic imbalance, leaving only the cells with the der(X) active (white). These latter cells functionally are genetically balanced, and the phenotype is normal. The individual has a skewed X-inactivation pattern.5 Or (d) the dark gray cells persist, in spite of their functional genetic imbalance (the defect is not severe enough to be lethal), and the individual is a mosaic of functionally balanced tissue (white cells) and imbalanced tissue (dark gray cells). In consequence, the phenotype is abnormal. (Adapted from Lanasa and Hogge, 2000). 5 An actual example of this sort of process, although occurring at a postnatal rather than an early embryonic stage of life, confined to one tissue (hematogenous), and at a mendelian rather than a chromosomal level, is given in Martinez-Pomar et al. (2005). These workers studied a girl with the syndrome of incontinentia pigmenti and immunodeficiency, due to the NEMO locus on the X chromosome. From age 2 to 4 years, her X-inactivation status in peripheral blood progressed from random to completely skewed in favor of the X with the normal NEMO gene, and in parallel there was correction of her immune function.
Sex Chromosome Translocations •
115
Measuring Inactivation Status. Inactivation status can be assessed cytogenetically (replication-banding, or R-banding), which enables, in principle, distinction of the early replicating (active) and the late replicating (inactivated) X chromosomes, and allows a precise estimate of the ratio of normal-Xactive to translocation-Xactive cells. Mostly, however, the analysis is done using molecular methodologies. The androgen receptor locus, at Xq13 (quite close to the XIC), is often used as the basis of this test. In the phenotypically normal heterozygote, the observation of a complete skew of translocation-Xactive and normal-Xactive, in the representative tissue analyzed, would indicate that the same 100:0 proportion applied elsewhere in the soma. Since it is impossible ever to test the entire soma (and in particular the brain), it would have to remain an open question, in a phenotypically abnormal but structurally balanced X-autosome heterozygote, that a more random skewing pattern might apply in some tissues, notwithstanding a complete skew in the peripheral tissue(s) tested. Abnormal individuals may show incomplete intertissue concordance of inactivation status, with sometimes quite different ratios in different tissues: say, 80:20 in blood and 30:70 in skin (Schmidt and Du Sart, 1992).
OVARI AN FUN C TIO N A ND THE X “C RITIC AL REGI ON S”
Breakpoints at certain locations in the X may affect ovarian function (Table 6–1). A breakage and reunion within either of two “critical regions” (CR1 and CR22) is characteristically associated with premature ovarian failure. CR1 is located in Xq21.1, and CR2 in Xq23-q28 (Fig. 6–1) (Rizzolio et al., 2006). In the case of Xq21 breakpoints, these may lead to an epigenetic down-regulation of “ovarian genes” located on the translocated autosomal segment, whereas breaks in Xq23-q28 may affect genes in the vicinity that normally require disomic expression (Rizzolio et al., 2009). In one series of 30 women presenting with premature ovarian failure, in whom the cytogenetic findings were reviewed, Devi and Benn (1999) recorded just one to be an X-autosome translocation heterozygote; thus, it is an infrequent cause of this problem.
Table 6–1. Occurrence of Gonadal Dysgenesis (Primary or Secondary) in 118 t(X-Autosome) Women According to X Chromosome Breakpoint
BREAKPOINT
Xpter-q12 q13 q13-q22 q22 q22-q25 q26 q27-qter
•
5 4 20 11 7 3 1
NORMAL GONADAL FUNCTION
37 8 1 6 1 5 9
Source: From Therman et al. (1990).
The Male X-Autosomal Hemizygote Almost invariably, the cytogenetically balanced male hemizygote is, without intervention, infertile, due to spermatogenic arrest; disruption of the sex vesicle (see later discussion) is the presumed proximate cause of the obstruction (Hwang et al., 2007). In two men subject to testicular biopsy, Quack et al. (1988) showed germ cell maturation arrest mostly at the pachytene stage of meiosis I, although a few cells managed to make the first and some even the second meiotic metaphase, and thus might have become spermatozoa. This outcome of a very modest success might more likely be achieved in those men in whom the breakpoints are more centromerically placed. A man hemizygous for a wholearm translocation (X;18)(q11;p11.1) was subject to sperm chromosomal fluorescence in situ hybridization (FISH) analysis; he had presented with infertility and “very severe oligoasthenoteratozoöspermia.” Analysis showed a range of segregant types in the small number of 447 cells able to be studied: alternate segregation in just over 50% (with half of these normal 23,Y), and adjacent-1, adjacent-2, 3:1, and 4:0 in 8%, 5%, 22%, and 2%, respectively (Perrin et al., 2008). If sperm can be retrieved, intracytoplasmic sperm injection (ICSI) may be attempted
2 Sometimes referred to as POF1 and POF2, for premature ovarian failure regions.
116
GONADAL DYSGENESIS
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
in the carrier male in order to enable fertility (see later).
Patterns of Inactivation in the Unbalanced Offspring FEMALE OFFSPRING OF THE X-AUTOS O MA L H ETEROZYGOT E OR HEMIZ YG OTE
As a rule (but one that can be broken), the pattern of inactivation that is observed, following selection, will be the one that allows the least amount of functional imbalance. This is typically arrived at in the karyotypically unbalanced daughter by inactivation of the abnormal chromosome, always supposing that the choice exists (and the choice can exist only if the abnormal chromosome contains an XIC). If the abnormal chromosome is a der(X) from a single-segment exchange, containing no autosomal material other than a telomeric tip, it comprises, essentially, a deleted Xp or Xq chromosome. In a girl with the 46,X,der(X) karyotype, preferential inactivation of this deleted X leads simply to a phenotype of partial Turner syndrome. Consider the family segregating a t(X;12) shown in Figures 6–2 and 6–4. The segregation shown in Figure 6–2 (daughter from adjacent-1) and Figure 6–4a (daughter) illustrates the case for an Xq deletion. Here, the normal X is active (shown as solid outline in Fig. 6–2), and the der(X) inactivated (dotted outline). Leichtman et al. (1978) provide an example of the Xp deletion circumstance in a three-generation family with seven persons having an Xp–Turner syndrome variant on the basis of a segregating t(X;1). If the der(X) carries a larger translocated autosomal segment—conferring, therefore, a partial autosomal trisomy in the 46,X,der(X) subject—the effects of this imbalance may be mitigated by selective inactivation of the abnormal chromosome. Transcriptional silencing can spread, albeit patchily, into the autosomal chromatin on the der(X), converting, at least partially, a structural autosomal trisomy into a functional autosomal disomy. Consider these two cases in which a practically complete trisomy 15—which typically causes first-trimester abortion (Rajcan-Separovic et al., 2001)—produced, in comparison, a very much attenuated phenotype. Garcia-Heras et al. (1998) reported on the terminated pregnancy of a t(X;15)(p22.2;q11.2) carrier mother, from whom the 19-week fetus with the der(X) was trisomic for 15q11.2-qter, but only
rather mild abnormalities of fetal morphology were to be noted. A t(X;15)(q22;q11.2) involving the same 15q11.2 breakpoint (in this case de novo) and diagnosed in a mildly dysmorphic and moderately developmentally delayed 3-year-old child, is described in Stankiewicz et al. (2006). A number of similar cases are on record, and the degree to which a phenotype may be attenuated is presumably related to the extent that genes in the autosomal segment are inactivated. Giorda et al. (2008) analyzed cells from a girl with mild dysmorphology, arthritis, obesity, microcephaly, and mental and behavioral disability, who had the karyotype 46,X,der(X)t(X;5)(q22.1;q31)dn and was thus partially trisomic for the large segment 5q31–5qter. They were able to show that, of 17 interpretable genes tested in this translocated segment, 9 had been inactivated, while another 8 were active (as measured by methylation status). This inactivation did not “weaken” as it spread further into 5q31– 5qter segment, and indeed the autosomal gene closest to the Xq-5q breakpoint remained active; thus, some autosomal genes were susceptible, and some were resistant, to the spreading influence from the XIST of the der(X). Figure 6–9 shows an example of blocked spread of inactivation into the autosomal (16p) segment of an X;16 translocation: observe the der(X) in the lower row, with pale (inactivated) long arm and dark (active) short arm. The converse, whereby the process of spreading autosomal inactivation may be detrimental to the phenotype, by converting a functional disomy (or near disomy) into a functional monosomy, is rarely observed. The family illustrated in Figure 6–11 provides a possible example. At first sight, one might have expected only a Turner syndrome phenotype in the daughter with a 45,X,der(X),–22 karyotype, since the essential defect appeared to comprise an Xp deletion, with her total complement of 22q material being intact, or nearly so. However, a more severe clinical picture evolved, and this may have reflected, speculatively, a transcriptional silencing of some crucial 22q loci, notwithstanding the apparent block to inactivation at the breakpoint on cytogenetic study. This case is mentioned further later. If, in the female with a 46,XX,der(autosome) karyotype, the derivative chromosome has no XIC in its translocated X-segment, this cannot be inactivated, and a functional partial X disomy is the consequence (Sivak et al., 1994). Figure 6–5 demonstrates a functional disomy for a part of Xp Sex Chromosome Translocations •
117
(a)
(b)
FIGURE 6–4 (a) Mother with balanced X;12 translocation, showing two different segregant outcomes. Her daughter had presented with clinical Turner syndrome, in whom the karyotype was initially interpreted as del(X)(q22). Her son was subsequently studied, and he had a partial Klinefelter syndrome. (Case of J. A. Sullivan.) (b) The presumed pachytene configuration during gametogenesis in the mother (X chromatin, open; chromosome 12 chromatin, crosshatched; dot indicates X-inactivation center). Light arrows indicate movements of chromosomes to daughter cells in adjacent-1 segregation, as observed in the daughter with partial Turner syndrome. Heavy arrows show the tertiary trisomy combination seen in the son with partial Klinefelter syndrome. These two segregations are represented in b and c in Figure 6–7.
(Xp22.31-pter)3 in a chromosomally unbalanced daughter; in this instance, since the autosomal breakpoint is at the telomere, we assume there to be little or no effect from a 10q monosomy. Gustashaw et al. (1994) describe a similar case in which they could be sure the Xp imbalance was the sole cause of
the abnormal phenotype, since the autosomal breakpoint was in 13p and the loss of one acrocentric short arm has, of itself, no effect. Functional disomy of distal Xq, Xq28-qter, has been reported sufficiently often that a clear core phenotype can be described (Sanlaville et al., 2005).
3 And presumably a functional trisomy for the pseudoautosomal region within this segment.
118
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
FIGURE 6–5 Functional X disomy. (a) Mother with balanced X;10 translocation (above), and her daughter with a 46,XX,der(10) karyotype from adjacent-1 segregation (below). The translocation is t(X;10) (p22.31;q26.3). Dashed box on cartoon karyotype indicates preferentially inactivated chromosome; dot indicates X-inactivation center. The der(10) contains Xp material in the translocated segment, which cannot be inactivated, and so the daughter has functional X disomy. Since the 10q breakpoint is in the terminal band we may regard this as an effectively single-segment exchange, with the phenotype of severe mental deficit and minor dysmorphism due entirely to disomy for the small Xp22.31→pter segment. (Case of A. Ma and H. R. Slater.) (b) The presumed pachytene configuration during gametogenesis in the mother (X chromatin, open; chromosome 10 chromatin, crosshatched; dot indicates X-inactivation center). Arrows indicate movements of chromosomes to daughter cells in adjacent-1 segregation; heavy arrows show the combination observed in this family. This is essentially the segregation a shown in Figure 6–7.
MALE OFFSPRING OF THE FEMALE X-AUTOS O ME H ETEROZYGOT E
Analogous to the female, the male inheriting a der(autosome) is affected according to whether the X translocated segment does or does not contain an XIC. If the X-segment lacks an XIC, a functional X disomy ensues, with a severe phenotypic effect. If the X-segment contains an XIC, the X-segment is inactivated and, other things being equal, a Klinefelter-like phenotype might be expected. But this expectation might not be met, and a more severe clinical picture, whether due to incomplete inactivation, or to the effect of a concomitant autosomal deletion, could result. Balcı et al. (2007) report a
three-generation family with a t(X;19)(q11;p13.3): a normal grandmother and mother with the balanced translocation, and a severely retarded boy, physically somewhat resembling Prader-Willi syndrome, whose karyotype was 46,XY,der(19) t(X;19). Virtually the entire Xq—including the XIC—was present in disomic dose on the der(19). Its otherwise lethal effect was considerably adjusted by inactivation, but nevertheless the phenotype was a great deal more severe than “Klinefelter-like.” Origin of the X-Autosome Translocation. All de novo balanced X-autosomal translocations so far studied have been of paternal origin, which may reflect the availability in male meiosis of the Sex Chromosome Translocations •
119
X chromosome for exchange with other chromosomes; the X pairs with the Y only at the PAR1, and the rest of the chromosome is unsynapsed (Turner, 2007). In one well-analyzed example, Giacalone and Francke (1992) did a molecular dissection on a de novo t(X;4)(p21.2;q31.22) in a girl with Duchenne muscular dystrophy and proposed a format in which two GAAT sequences 5 kb apart in Xp, and one GAAT in 4q, came together during meiosis in spermatogenesis, deleted the 5 kb length in Xp (which comprised a small part of the dystrophin gene), and reformed as a der(X) and a der(4). Similar mechanisms likely underlie the formation of most X-autosome translocations. Once a balanced translocation is established in a family, and if the heterozygous state is associated with phenotypic normality, male infertility dictates that transmission thereafter will be matrilineal. We comment upon the de novo X-autosome translocation identified at prenatal diagnosis on p. 469. Translocations That Disrupt X-Borne Loci. Quite a number of X-linked disorders have been seen in females with (typically de novo) X-autosome translocations. The resulting clinical problems may manifest because of nonrandom X-inactivation, disruption of a gene, or a combination of both. With preferential inactivation of the normal X, there remains no functional copy of the normal allele: an effectively “nulliallelic” state. Among several other examples, we may point to the case of twin sisters with de novo balanced reciprocal translocations between Xp and 5q (Zenker et al., 2005). These girls had ornithine transcarbamylase (OTC) deficiency
due to a combination of disruption of the OTC gene and skewed inactivation of the normal X chromosome. The disease state resulted, with no enzyme produced, because one allele was disrupted and the other allele was silenced. X-autosome translocations have offered research potential, and some loci have been initially mapped by study of rare/unique female patients with the particular mendelian condition (Schlessinger et al., 1993). The classic historical case is the female Xp21-autosome translocation heterozygote who has Duchenne/Becker muscular dystrophy, as mentioned earlier, and an example of which is illustrated in Figure 6–6. The X chromosome may have a particular load of “brain genes” (Turner, 1996), and it is of interest that a number of pure brain-related phenotypes, without dysmorphism or malformation, have been associated with these translocations. A girl with lissencephaly and an X;2 translocation pointed the way to the discovery of the doublecortin gene at Xq22.3 (Gleeson et al., 1998). The disruption of an X-linked neuronal gene, oligophrenin-1, caused isolated mental defect in a female with an X-autosome translocation 46,X,t(X;12)(q11;q15). The breakpoint was in the second intron, and thus the first two exons of the gene were on the der(12), and the remaining 23 exons on the der(X). No transcript could be produced, due to this disruption of the allele, and with the other allele on the normal X having been inactivated (Billuart et al., 1998). Another gene at Xq11, with effects in a number of compartments of the neural substrate (but not outside it), is collybistin, which influences a specific type of neuronal receptor. A woman with an X;18
FIGURE 6–6 A de novo X-autosome translocation 46,X,t(X;4)(p21;p16) in which the dystrophin locus at the Xp21 breakpoint is presumed to be disrupted, in a 7-year-old girl. In consequence very little dystrophin is produced, and the girl has a Becker-like muscular dystrophy. The approximate position of the dystrophin locus is indicated (arrowhead) on the intact X. The intact X is preferentially inactivated, as shown here with replicationbanding and indicated in dashed outline on the cartoon karyotype. Early replicating (active) chromatin and the late replicating (inactivated) chromatin stain dark and light, respectively. (Case of J. A. Sullivan.) 120
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
translocation that disrupted collybistin (the breakpoint of the 18 was in a region devoid of genes) presented a syndrome of mental retardation, aggressive behavior, epilepsy, anxiety, and a disturbed sleep pattern (Kalscheuer et al., 2009). And rare patients with an X-linked dominant infantile spasm syndrome, reminiscent of the Rett syndrome phenotype, and having its basis in the gene CDKL5, have had translocations in which the X breakpoint is within the CDKL5 locus at p22.1 (Córdova-Fletes et al., 2010).
DETAILS OF MEIOTIC BEHAVIOR FEMALE MEIOSIS In oögenesis, a quadrivalent presumably forms, just as in the two-way translocation between autosomes. 2:2 alternate segregation with the intact X and intact autosome can lead to 46,XX or 46,XY conceptions, while transmission of the translocation in balanced state produces heterozygous or hemizygous conceptions. As for malsegregation, Figures 6–7 and 6–8 set out certain outcomes that may be viable, for various categories of single-segment and doublesegment translocation, as discussed later. Given the greater survivability of X imbalances due to inactivation, and likewise a possible lessened effect of autosomal imbalance, a greater number of conceptuses are potentially viable than from the autosomeautosome translocation. The “rules” of segregation (p. 77) may not apply; for example, a viable adjacent-2 malsegregation can occur with a derivative chromosome having a large centric segment. The coexistence of tertiary monosomy and adjacent-2 aneuploidy in the family described in Figure 6–11, two otherwise very uncommon segregations, reflects the unique characteristics of the X-autosome translocation.
Categories of Translocation and Modes of Malsegregation We consider here various chromosomal scenarios, which ought to cover the majority of clinical circumstances. Concerning terminology with respect to the size of translocated segments: if one of the translocation breakpoints is at the telomeric tip of either the autosome or the X chromosome, and thus only one of the translocated segments (X or autosomal) comprises an important amount of
chromatin, this may be considered an effective “single-segment exchange.” If both translocated segments are of significant size, this is a “doublesegment exchange.” SI NG L E-SEG M EN T EXCH A N G E, X T R A N S LO CAT ED SEG M EN T
The first two columns in Figure 6–7 and the first column in Figure 6–8, segregations a–c and segregation a, respectively, depict the general form of a translocation in which the single important exchanged segment comprises X chromatin. A particular example is shown in Figure 6–4, in which the derivative X chromosome is deleted for a large segment of Xq and has only the telomeric tip of 12p in exchange. A child receiving this abnormal “Xq–” in place of a normal X, or as an additional chromosome, could present with a partial form of a sex chromosome aneuploidy syndrome. Thus, a daughter with 46,X,der(X) from adjacent-1 malsegregation (b in Fig. 6–7) would have a variant form of Turner syndrome. From tertiary trisomy (c in Fig. 6–7), a son with 47,XY,+der(X) would have incomplete Klinefelter syndrome; and a 47,XX,+der(X) daughter might show the 47,XXX phenotype to a diminished degree. More severe consequences follow the countertype adjacent-1 segregation, a in Figure 6–7. Conceptions with 46,der(12) from adjacent-1 segregation would, in the family in Figure 6–4, be functionally disomic for a large, unsurvivable amount of Xq, and they would abort. However, if the translocated X segment is small, the functionally disomic X state may be viable. This is shown in Figure 6–5, in which the mentally retarded and dysmorphic daughter has a 46,XX,der(10) karyotype and is functionally disomic for the small amount of Xp22.31-pter. As for adjacent-2 segregation (Fig. 6–8a), such a gamete would, in theory, have viability only if it is the der(autosome) that is transmitted, along with the intact autosome, and if the X segment of the der(autosome) includes the XIC. In that case, inactivation could spread through the autosomal material, converting, at least partially, a structural autosomal trisomy into a functional autosomal disomy. Of course, there would be a partial X monosomy as well. This scenario is discussed in more detail in the section on “Double-Segment Exchange, Adjacent-2.” A truly single-segment X-autosome translocation, the translocated segment comprising Sex Chromosome Translocations •
121
FIGURE 6–7 Major categories of adjacent-1 and 3:1 malsegregation in the X-autosome female carrier. The top row shows quadrivalents at maternal meiosis, and the following rows various combinations of segregant products. Open, X chromatin; crosshatched, autosomal chromatin; dot indicates X-inactivation center. “Single-segment” and “double-segment” are defined in the text. X exchanges can occur in either Xp or Xq; only Xq exchanges shown here. Circled letters provide reference points for text comments. *Effect of autosomal duplication may be lessened by spreading of transcriptional silencing into the autosomal segment of the der(X). **Blocking of spread of inactivation into the autosomal segment of the der(X) may avoid further functional autosomal monosomic effect.
FIGURE 6–8 Three categories of adjacent-2 malsegregation in the X-autosome female carrier. The top row shows quadrivalents at maternal meiosis, and the next row various combinations of adjacent-2 segregant products. Note that these potentially viable outcomes occur only in the setting of the transmitted derivative chromosome, be it the der(X) or the der(autosome), having an X-inactivation center (XIC). In the first two columns, the der(autosome) has the XIC; here, the X breakpoint must be in proximal Xq, above the XIC, as depicted. In the third column, in which the der(X) has the XIC, X exchanges can occur either in Xp, or in Xq distal to the XIC; only an Xp exchange is shown here. Open, X chromatin; crosshatched, autosomal chromatin; dot indicates XIC; der(A), der(autosome). Circled letters provide reference points for text comments. *Effect of autosomal duplication may be lessened by spreading of transcriptional silencing into the autosomal segment of the der(A). **Blocking of spread of inactivation into the autosomal segment of the der(X) may avoid further functional autosomal monosomic effect.
X material, is recorded in de Vries et al. (1999). A mother had a submicroscopic segment of the PAR1 in distal Xp (p22.31-pter) translocated across to the short arm of a chromosome 14, but, as far as could be seen, there was no reciprocal movement back to the X of any 14p material. She transmitted the der(X) to a son, who presented signs interpreted as consistent with nullisomy for certain genes in the distal PAR1: the SHOX, MRX, CDPX, and STS genes, their absences responsible respectively and collectively for short stature,
developmental delay, short limbs, and ichthyosis. SI NG L E-SEG M EN T EXCH A N G E, AU TOS O M A L T R A N S LO CAT ED SEG M EN T
The single segment being of autosomal origin, with only the telomeric tip of Xp or Xq translocated in exchange, is shown in the middle column of Figure 6–7, segregations d–g. The imbalanced conceptions from 2:2 adjacent-1 malsegregation would be partially monosomic or partially trisomic for the Sex Chromosome Translocations •
123
autosomal segment: 46,der(autosome) and 46,der(X), respectively (segregations d and e in Fig. 6–7). The partial autosomal trisomic state may, in the 46,X,der(X) female, have an attenuated phenotype due to spreading of inactivation from the XIC of the der(X) into the autosomal segment. The 46,Y,der(X) male conceptus, in which no X-inactivation occurs, would show the undiluted effect of the partial autosomal trisomy. The partially monosomic state, 46,XX,der(autosome) or 46,XY,der(autosome), would be no different than if the other chromosome participating in the translocation had been an autosome, instead of an X, and the typical clinical consequence associated with that autosomal deletion would be expected. D OU B LE-SEG MEN T E XC HA NGE , ADJACENT-1
In a double-segment exchange with adjacent-1 segregation (right column, Fig. 6–7, segregations h–i), there may be, in the unbalanced conceptus, effects of a combined X functional disomy and autosomal monosomy, or of X monosomy (or nullisomy) and autosomal trisomy. Such combinations would often be lethal in utero. But in the 46,X,der(X) female (segregation i), the effects may be very considerably modified by spreading of inactivation. Consider the t(X;16) illustrated in Figure 6–9. The 46,X,der(X) daughter has both a monosomy for most of Xp, giving a Turner-like phenotype, and a structural trisomy for most of 16p. Following spread of inactivation in the der(X) into its autosomal segment in a fraction of cells, the 16p trisomy has been converted, in these cells, into a functional 16p disomy. In 76% of cells, however (and in the cell illustrated), the inactivation has not extended into the 16p segment. Thus, she has, effectively, a functional mosaic 16p trisomy/16p disomy. This same combination with a Y replacing the X as the intact sex chromosome, 46,Y,der(X), with nullisomy Xp/trisomy 16p, would not be viable. The other adjacent-1 conceptions with 46,XX,der(16) and 46,XY,der(16) (light arrows in Fig. 6–9; h in Fig. 6–7) would not be similarly “modifiable” and would have a very large functional imbalance, and they would also be expected to abort early in the pregnancy. D OU B LE-SEG MEN T E XC HA NGE , ADJACENT-2
Adjacent-2 segregation typically produces trisomy for much of one chromosome along with monosomy 124
•
for much of the other, and this is not, in the usual autosome-autosome translocation, remotely viable (e.g., segregation (5) in Fig. 5–4). But such an enormous degree of structural imbalance can be accommodated in some X-autosome translocations, in a female conceptus. First, consider the case of the intact autosome and the derivative autosome being transmitted together: 46,X,–X,+der(autosome). Provided the X segment of the der(autosome) includes the XIC (segregation b in Fig. 6–8), inactivation can spread from the XIC in both directions and into the autosomal segment, counteracting the effect of the autosomal duplication, at least partially. The concomitant partial X monosomy is, of itself, a viable state. The child would be expected to display a partial Turner phenotype, upon which the effect of a variably inactivated partial autosomal trisomy would be added. This is illustrated in Leisti et al. (1975), who record a mother carrying a t(X;9) (q11;q32) and her daughter being 46,X,–X,+der(9). In the daughter, inactivation spread through much of the autosomal segment, which very substantially, although not completely, neutralized the effect of the partial trisomy 9: she had a Turner phenotype with superadded microcephaly and mental defect. The case in Williams and Dear (1987) is similar, with a retarded and dysmorphic child having the karyotype 46,X,–X,+der(10),t(X;10)(q11;q25) mat, but in this instance inactivation into the autosomal segment was apparently blocked at the centromere of the der(10). This left the child with an effective duplication of 10p, along with the X deletion (Fig. 6–10). Concerning a male conception in this setting, of course an adjacent-2 conceptus could not survive. Second, viability is also possible in one very rare circumstance of an intact X and the der(X) being transmitted together, with the adjacent-2 karyotype 46,XX,–(autosome),+der(X), segregation c in Figure 6–8. The der(X) must contain an XIC; its autosomal segment must comprise a very substantial amount of the chromatin of that autosome; and there must be little or no spread of inactivation beyond the X segment of the translocation chromosome into the autosomal segment. In this way, the autosomal component can maintain sufficient disomic genetic activity to produce a viable phenotype. Only autosomes with “genetically small” short arms could enable these criteria to be met. An example from a maternal t(X;22)(p21.3;q11.21) is shown in Figure 6–11 (daughter with adjacent-2, bottom row). The der(X) comprises most of an X and all, or
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
FIGURE 6–9 Spread of inactivation into autosomal segment. (a) Mother with balanced X;16 translocation (above), and her daughter with a 46,X,der(X) karyotype from adjacent-1 segregation (below). The translocation is t(X;16)(p11;p12). Replication-banding shows active (darker-staining) and inactive (lighter-staining) chromosome segments. The normal X is inactivated in all cells analyzed in the mother (dashed box on cartoon karyotype; dot indicates X-inactivation center). The daughter’s abnormal X lacks Xp and contains distal 16p material. This chromosome is preferentially inactivated (dashed outline of box), but in 76% of cells analyzed (lymphocytes) the inactivation has not continued through the translocated 16p segment (dotted outline of box). The phenotype is the combined result of the Xp monosomy and a “partial” 16p trisomy. The child is short and has a developmental age of about 2½ at a chronological age of 4 years. (Case of C. E. Vaux.) One other daughter had the same balanced translocation as the mother and showed consistent inactivation of the normal X chromosome in blood lymphocytes, but she suffered intellectual deficit. (b) The presumed pachytene configuration during gametogenesis in the mother (X chromatin, open; chromosome 16 chromatin, crosshatched; dot indicates X-inactivation center). Arrows indicate movements of chromosomes to daughter cells in adjacent-1 segregation; heavy arrows show the combination observed in this family. This is essentially the segregation i in Figure 6–7, with an Xp breakpoint in this case.
(a)
(b)
FIGURE 6–10 Adjacent-2 segregation. (a) Mother with balanced X;10 translocation (above), and her daughter with a 46,X,–X,+der(10) karyotype (below), on G-banding. The translocation is t(X;10)(q11;q25). Replication-banding showed the normal X to be inactivated in all 30 lymphocytes analyzed in the mother (dashed box on cartoon karyotype; dot indicates X-inactivation center). The daughter’s der(10) was preferentially inactivated (dashed outline of box) in 50/50 cells, but the inactivation did not continue through to the 10p segment (dotted outline of box). The phenotype is the combined result of the 10p duplication and Xp monosomy. (Case of J. Williams; Williams and Dear, 1987.) (b) The presumed pachytene configuration during gametogenesis in the mother (X chromatin, open; chromosome 10 chromatin, crosshatched; dot indicates X-inactivation center). Arrows indicate movements of chromosomes to daughter cells in adjacent-2 segregation; heavy arrows show the combination observed in this family. This is segregation b in Figure 6–8.
almost all, of 22q. If its 22q segment were blocked from inactivation, there would be, in effect, a nearnormal functional disomy 22, along with a partial XXX syndrome. In fact, this woman had a mild intellectual disability and attended a special school; the relative contributions to her phenotype of the two components of the cytogenetic abnormality are open to speculation. 126
•
DOUB L E-SEG M EN T EXCH A N G E, 3 : 1 SEG R EG AT I O N W I T H T ERT I A R Y M O N OS O MY
The same criteria noted in the earlier paragraph may also obtain in the rare situation of tertiary monosomy being viable (in essence, this is segregation k in Figure 6–7). The t(X;22) in Figure 6–11 again
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
(c)
FIGURE 6–11 3:1 tertiary monosomy and adjacent-2 segregation both occurring in the same family (and see text). (a) Pedigree of family segregating a t(X;22)(p21.3;q11.21). Filled symbol, imbalanced state; half-filled symbols, heterozygote/hemizygote; N = 46,XX. (b) Partial karyotypes of heterozygotes (top), and of the two unbalanced states (lower). On replication-banding, the normal X is inactivated in all cells analyzed in the heterozygotes, whereas the der(X) is inactivated in the two affected persons (dashed box on cartoon karyotype; dot indicates X-inactivation center). In the affected child in generation III with a 45,X,–X,+der(X),–22 karyotype (middle karyotype), the der(X) was positive for a probe recognizing a sequence in the DiGeorge critical region. The der(X) chromosome showed, in 50/50 cells, apparently no inactivation going through to its 22 component (dotted outline of box), but the clinical picture might suggest otherwise (see text). The affected woman II:1 has the adjacent-2 karyotype 46,XX,+der(X),–22. (Case of T. Burgess.) (c) The presumed pachytene configuration during gametogenesis in the heterozygote (X chromatin, open; chromosome 22 chromatin, crosshatched). Heavy arrow indicates movement of the der(X) chromosome to one daughter cell in 3:1 segregation (essentially segregation k, Fig. 6–7). Dashed arrows show the movement of chromosomes in the adjacent-2 combination (segregation c, Fig. 6–8).
provides an example. In the index case in this family having the tertiary monosomy state 45,X,– X,+der(X),–22 (middle row, Fig. 6–11), the der(X) chromosome is preferentially inactivated, but inactivation has not (at least on blood lymphocytes) spread through to the 22q component of the der(X). Thus, a functional 22 disomy is maintained, or nearly so. The important structural imbalance, one might have predicted, could have been limited to the Xp21.3-pter deletion (loss of 22p being without effect), with the phenotype confined to a Turnerlike picture. In the event, however, there were minor congenital anomalies, and she was assessed as being intellectually disabled. This does suggest that the pattern of inactivation elsewhere in the soma may have differed from that observed on peripheral blood, and there might have been a degree of functional 22q monosomy in other tissues, including brain.
homologous regions on the der(X) and the der(A) (Fig. 6–12). As with the female, some malsegregant forms would have potential viability, due to the potential lesser effects of X imbalance. Similarly according to the principles as set out earlier for the female, but with the additional factor of a Y chromosome to be considered, the reader can determine the range of possibilities in a particular case, for this rarely encountered circumstance.
(a)
3:1 SEG R EG ATIO N , I N T E RCHA N GE TRI SO MY / MO N OS O MY
From each of the categories of single- and doublesegment exchange, 3:1 interchange trisomy could theoretically produce Klinefelter syndrome or XXX syndrome along with the balanced translocation; and interchange monosomy could produce standard 45,X Turner syndrome. We are aware of only one such outcome, an infertile woman with 47,XX,t(X;12) from a 46,X,t(X;12)(q22;p12) mother, the imbalance being equivalent to 47,XXX (Madan et al., 1981). 4:0 SEG R EG ATIO N
With a trisomically viable autosome, say chromosome 21, a 48,XX,+der(X),+der(21) karyotype might be equivalent to the potentially viable 48,XXX,+21 state, a combined Down syndrome plus XXX syndrome. We know of no such report.
MALE MEIOSIS Meiosis in the X-autosome hemizygote is typically compromised, due to failure of formation of the sex vesicle, and spermatogenesis arrests. Infrequently, some sperm may be made, albeit in small numbers. Perrin et al. (2008) propose formation of a quadrivalent, in which the Y chromosome participates with apposition of its PAR1 and PAR2 to the
128
•
der(X)
Y
18
der(18)
(b) der(X) 18
der(18)
Y
FIGURE 6–12 The X-autosome translocation in the carrier male, if meiosis is able to proceed. (a) Father with balanced X;18 translocation, from whom pregnancy was achieved following in vitro fertilization with intracytoplasmic sperm injection. (b) The presumed pachytene configuration during gametogenesis in the father (X chromatin, open; Y chromatin, gray; chromosome 18 chromatin, crosshatched). According to this construction, the X segments and the Y align only at the respective pseudoautosomal regions (PARs), and otherwise lie free. Heavy arrows indicate movement of chromosomes to daughter cells in one of the 2:2 alternate segregations, to produce a normal gamete, as observed in the 46,XY son. Light arrows show the other alternate combination, which could lead to a carrier daughter. (Drawn after the case in Perrin et al. 2008.)
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Y-AUTOSOME TRANSLOCATIONS Y-autosome translocations fall into two major Yq-breakpoint categories, one of which has important clinical implications, and the other of which does not. Certain other rare forms exist. First, some brief comments on the nature of the Y chromosome are in order. THE RO LE A N D B EH AV I OR OF THE Y C H RO MOS O M E
The particular raison d’être of the Y chromosome is to bring about male development. The testisdetermining gene, SRY, lies in the euchromatic region on the short arm, just 5 kb proximal to the pseudoautosomal boundary. As noted earlier, and see Figure 6–1, the primary pseudoautosomal regions (PAR1) of the Y and X short arm contain homologous loci, and certain other loci elsewhere in the Y have homologs on the X (Disteche, 1995). The secondary pseudoautosomal region (PAR2) is located at distal Yq and distal Xq; its loss from Yq seems to be without phenotypic consequence (Kühl et al., 2001). From the point of view of reproductive health, three “azoöspermia factor regions” on Yq are of importance, named AZFa, b, and c, and these are discussed in more detail in Chapter 23. Otherwise, about half the Y—the amount is variable—comprises the genetically inert heterochromatic region of the long arm (Yq12), which contains highly repetitive DNA sequences.
MEIOSIS During normal meiosis in the male, the X and Y chromosomes recombine, synapsing at the PAR1 at the tips of Xp and Yp. The two sex chromosomes joined together in this way, but otherwise unsynapsed, comprise the “sex vesicle” (or sex-body, or XY-body), which can be considered as a peripheral nuclear subdomain within which the X and Y chromosomes lie, genetically inactivated, during the pachytene stage of meiosis (Turner, 2007). A properly formed sex vesicle is necessary for normal spermatogenesis, and anything that interferes with its normal formation—such as the presence of a translocation chromosome—will compromise the process of sperm development. We have seen earlier
that an X-autosome translocation in the male practically always causes spermatogenic arrest. And fairly infrequently, some autosomal translocations, and more especially those involving an acrocentric chromosome, can cause interference with the sex vesicle, with consequential infertility. Similar considerations apply to the balanced Y-autosome translocation (other than the Y-acrocentric translocation) (Maraschio et al., 1994). The autosomal components of the quadrivalent, “dragged into” the sex vesicle, as it were, can have a damaging effect, possibly due to their being inactivated, and preventing any further progress of meiosis (Turner, 2007). Delobel et al. (1998) illustrate this circumstance in their study of a phenotypically normal infertile man with a translocation t(Y;6)(q12; p11.1). They analyzed meiotic preparations from a testicular biopsy, noting the configuration of the quadrivalent, this comprising the X,der(Y),der(6), and normal chromosome 6. The autosomal elements of the quadrivalent were seen to have been drawn into the sex vesicle and to be hypercondensed. The result was spermatogenic arrest at the pachytene stage, with subsequent degeneration of spermatocytes. In a similar case, Sun et al. (2005) analyzed testicular tissue from an azoöspermic man with a Yq;1q translocation and showed (using MLH1 staining) a reduced level of meiotic recombination, with only a small fraction of cells progressing through to pachytene, and again the autosomal translocated segment drawn into the sex vesicle. Yet (and in contradistinction to the X-autosome story in the male) the Y-autosome carrier may occasionally retain natural fertility. Otherwise, fertility may be “rescueable” by means of assisted reproduction, as discussed further later.
Balanced Reciprocal Yq and Autosome4 Translocation Reciprocal exchange between the Y long arm and an autosome produces a balanced Y-autosome translocation. In the form being considered here, the Y breakpoint is usually given as q11.2 or q12, and the autosomal breakpoint is anywhere on the autosomal karyotype, other than an acrocentric short arm (Braun-Falco et al., 2007). There are associated phenotypic abnormalities in a few individuals, and this may be due to a disruptive effect at the breakpoints, or a deletion of autosomal material distal to the
4 Other than an acrocentric short arm.
Sex Chromosome Translocations •
129
breakpoint (Erickson et al., 1995). In most, the rearrangement may be truly balanced, with the physical and intellectual phenotype being normal, and infertility is the usual presenting factor. Given this latter fact, it follows logically enough that the translocation would typically arise as a de novo event, and this is indeed the observation (Pinho et al., 2005). The infertility may be a result of disruption of the sex vesicle, as discussed earlier. In other cases, documented loss of the AZF region may explain the infertility (Brisset et al., 2005). If spermatogenesis is able to proceed, there is a risk for the generation of unbalanced forms, and a few examples are on record. Mademont-Soler et al. (2009) describe a fertile couple, the father having the karyotype 46,X,t(Y;12)(q12;q24.33). The der(12) was deleted for a very small distal segment of chromosome 12 (12q24.33-qter), whereas the der(Y) carried this material. In two pregnancies, the two different adjacent-1 segregations were observed: 46,der(12) in one, and 46,der(Y) in the other. Both pregnancies, with thus an autosomal deletion and a duplication respectively, were terminated. A few familial cases have been reported, with father and son having the same balanced rearrangement. Teyssier et al. (1993) document a man with severe oligoasthenospermia who had a t(Y;1) (q11;q11), and whose father proved to carry the same translocation. Intact fertility is well illustrated in the family described by Sklower Brooks et al. (1998), depicted in Figure 6–13. One son in a sibship of five males and two females, he himself a university graduate, had presented for genetic counseling when his wife had a third miscarriage (they also had a normal daughter). The deceased father must have carried a t(Y;8)(q12;p21.3), with three sons showing the balanced state and two sons having inherited an unbalanced complement, 46,X,der(Y). The unbalanced state conferred a partial trisomy for 8p22-pter, which was associated with a mild learning difficulty. With access to ICSI (intractyoplasmic sperm injection), biological paternity becomes possible for some carriers. The man with the 46,X,t(Y;18) (q11.2q21) translocation shown in Figure 6–14 had been karyotyped in the course of investigation for infertility with severe oligospermia, he being an otherwise normal person. In this Y;18 case, of the 16 possible embryos, more than half, including one of the 4:0 segregants, would in theory be viable; the reader may wish to work out which ones these might be. Only one sperm, the 23,X, is capable of 130
•
producing a phenotypically and karyotypically normal child; the other gamete from alternate segregation, 23,t(Y;18), would produce a son who would likely have similar infertility. At preimplantation genetic diagnosis (PGD), the chromosomally unbalanced embryos could be discarded. With a small number of eggs retrieved on each stimulation cycle, a normal combination might well not happen, given that there are 14 unbalanced possibilities, if each outcome were equally likely. But in fact the observations of Sklower Brooks et al. (see earlier discussion) and Giltay et al. (see later discussion) provide some encouragement that the odds for the Y-autosome carrier (in other words, the meiotic predisposition) may be tipped in favor of the normal and balanced forms. As it turned out in this Y;18 case, one embryo was indeed 46,XX, and this was successfully implanted. This question is more directly answered in Giltay et al. (1999), these workers undertaking a sperm analysis in a man with a t(Y;16)(q11.21;q24). Sperm were present, but few in number, with many abnormal forms (oligoasthenoteratozoöspermia). Although alternate segregation accounts for only 2 out of the 16 segregation possibilities, in fact in this case half of all morphologically normal spermatozoa were normal or balanced, with about 40% showing adjacent segregation, and about 10% with 3:1. But the fractions were less favorable if abnormal sperm were included. With reference to assisted conception and using ICSI, Giltay et al. speak of in vitro and in vivo selection (the former artificial, the latter natural) combining to effect a considerable reduction in the risk for an unbalanced offspring. In fact, this man had had three children by ICSI, two normal daughters and a carrier son.
Y Long-Arm and Acrocentric Short-Arm Translocations Yqh MATERIAL ON ACROCENTRIC SHORT A RM , 4 6 - CH R O M OS O M E CO UN T
This is the most common form of Y-autosome translocation, accounting for some 70% of cases. The autosome is one of the acrocentrics, most commonly chromosome 15, followed by chromosome 22. There is no loss or gain of euchromatin; the result is that one acrocentric carries some phenotypically irrelevant Y heterochromatin, looking rather like (and sometimes mistaken for) a very long short arm (Neumann et al., 1992). The breakpoints
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
(c)
FIGURE 6–13 A Y-autosome translocation, not involving an acrocentric short arm. In this particular example, and somewhat unusually, fertility is apparently normal. The autosomal translocated segment is of small size, structurally and functionally, and the aneuploid state with a dup(8p) is not only viable, but associated with only a mildly abnormal intellectual phenotype and an essentially normal physical appearance. (a) Family tree. Filled symbol, unbalanced karyotype; half-filled symbol, balanced carrier. The deceased grandfather is presumed to have been a translocation heterozygote. (b) Partial karyotype of a translocation heterozygote (above), showing the Y;8 translocation, and one of the individuals with the unbalanced complement (below). (c) The presumed pachytene configuration during gametogenesis in the heterozygote (chromosome 8 chromatin, open; Y and X chromatin, crosshatched). Arrows indicate movements of chromosomes to daughter cells in “adjacent-1” segregation; heavy arrows show the combination observed in this family. (From S. Sklower Brooks et al., 1998, Normal adaptive function with learning disability in duplication 8p including band p22, American Journal of Medical Genetics 78:114–117, and with the permission of Wiley-Liss.)
are sited in the acrocentric short arm (p11-p13) and in the heterochromatin of the Y long arm (Yq12) (Fig. 6–15). Males and females can equally be carriers. Neither phenotypic abnormality nor infertility appears to be associated (Hsu, 1994). Rare disquieting reports need to be viewed cautiously, such as
that of Rajcan-Separovic et al. (2001), who raise the possibility of a secondary chromosomal abnormality, in documenting the remarkable instance of a woman with the karyotype 46,XX,der(15)t(Y;15) (q12; p13)mat, of older childbearing age, who had had two trisomic 15 pregnancies. Sex Chromosome Translocations •
131
Father
18 der(18) der(Y) X
18 der(18) der(Y) X
Potential daughter
18
18
X
X
18
18
X
X
FIGURE 6–14 The Y-autosome translocation and infertility. This t(Y;18)(q11;2q21) was identified in a man presenting with oligospermia during investigation for infertility. The fact that some sperm are still being produced allows the option of in vitro fertilization with intracytoplasmic sperm injection. A considerable number of these unbalanced gametes could, in theory, be viable. Only a 46,XX daughter could be both karyotypically and phenotypically normal. (Case of L. Harris and L. Wilton.)
Rare Forms Yqh MATERIAL ON A NONACROCENTRIC CHRO MOS O ME, 4 6 - C HROM OSOM E COU NT
If Yq heterochromatin is translocated to the tip of an autosome, other than to an acrocentric short arm, there may or may not be reproductive implications.
FIGURE 6–15 An example of the Y-autosome translocation involving an acrocentric autosome, the der(15) depicted here, with the breakpoint in the acrocentric short arm. Normal chromosome 15 and normal Y shown alongside for comparison (chromosome 15 chromatin, crosshatched; Y heterochromatin, filled; Y euchromatin, open). The translocation chromosome can be carried equally by males and females. The karyotype appears unbalanced, but the phenotype is normal. 132
•
A der(1)t(Y;1)(q12;p36) in a French family could be traced back to a common couple married in 1773, with self-evident fertility, male and female, for more than two centuries (Morel et al., 2002a), and Vozdova et al. (2011) showed normal seminal indices in a man with a familial der(4)t(Y;4) (q11.23;p16.3). Y MATERIAL PLUS ACROCENTRIC SHORT A RM T I P, 4 6 - CH R O M OS O M E CO UN T
This is essentially the countertype of the common case exemplified by the Y;15 described earlier, in which the other reciprocal product, the der(Y), replaces a normal Y. Hoshi et al. (1998) identified a perfectly normal man, the father of three, who had a 46,X,–Y,t(Y;15)(q12;p13) karyotype. The der(Y) contained the necessary male-determining and fertility regions. He was only investigated because his sister had a gonadal tumor of testicular origin, she having the mosaic karyotype 46,X,Y,t(Y;15)/45,X. Y Material to Autosome Translocation, 45-Chromosome Count, Including “45,X Male”. The testis-determining region of the Y, containing the SRY gene, can be translocated onto an autosome, usually an acrocentric (Farah et al., 1994). The individual, phenotypically male, has 45 chromosomes, including the Y+autosome fusion product. The translocated Y segment may be beyond the level of cytogenetic resolution, and the karyotype can appear as 45,X (“45,X male”) until further studies cast light (C-P Chen et al., 2008). The translocation may be of no phenotypic or reproductive effect, as Callen et al. (1987) record in a family identified quite by chance, in the course of a research study, in which a man and two sons had the karyotype 45,X,dic(Y;22)(q11.23;p11.2). A similar story is presented in Morales et al. (2007), in this case the karyotype being 45,X,psu dic(Y;22)(qter;p11.2), with the Y+22 chromosome comprising almost an entire Y and almost an entire 22. The chromosome very evidently did not affect fertility in 10 carrier fathers in this large family; this may have reflected that recombination of the PAR1 of the Y with that of the X was not obstructed, due to the chromosome 22 component being well “out of the way” of the sex vesicle, so to speak. The active centromere of the Y+22 chromosome was from the chromosome 22, the Y centromere being inactivated, and thus it was segregation of the 22 chromosomes that was the sexdetermining mechanism in the offspring of these
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
men. It was interesting that only normal children were born, whereas, in theory, segregation might also have led to 45,X Turner syndrome and 46,XX,psu dic(Y;22) Klinefelter syndrome. More often, the reproductive and sometimes the physical phenotype is affected. Azoöspermia is a frequent finding, as documented in a review of 31 cases of “45,X male” (C-P Chen et al., 2008). The Y component may be translocated insertionally, as Yenamandra et al. (1997) demonstrated in a phenotypically abnormal “45,X” boy, in one of whose chromosomes 4, at 4p15.3, the SRY-bearing segment was accommodated. Rather more obvious cytogenetically was the de novo dicentric Y;13 translocation, 45,X,dic(Y;13)(p11.3;p11.2), described in Shanske et al. (1999a): the translocation comprised almost a complete 13+Y composite. Their patient was a very short and otherwise normal 10-year-old boy, in whom the SHOX growth control gene, normally located in the PAR1, was absent. A patient with a very similar karyotype reported in Alves et al. (2002) was a man, 170 cm tall, with severe oligospermia. Yp;15q Translocation and Prader-Willi Syndrome. A very few cases of Prader-Willi syndrome (PWS) have been due to a fusion between a Y and a chromosome 15, having the karyotype 45,X,t(Y;15) with varying breakpoints, either de novo or familial (Vickers et al., 1994; Puvabanditsin et al., 2007). A remarkable example concerns a familial t(Y;15)(p11.2;q12), described in Gole et al. (2004). The father with the balanced translocation had a daughter with presumed PWS, she having inherited his X chromosome and the der(Y) which comprised almost all of 15q but lacking the PWS region, and most of the Y but lacking SRY. Her karyotype 46,XX,–15,+der(Y) reflected what might be called a “version” of adjacent-2 segregation. The absence of a paternally originating PWS region led to the development of that syndrome, while the absence of SRY was the basis of female sex. Her brother had the countertype “standard” adjacent-2 combination, 46,X,–Y,+der(15): he was of below-average intelligence, presumably due to duplication of the proximal 15q region, 15cen→q12, and had required treatment for hypogonadism The De Novo Yq;1q Translocation with 1q Trisomy. Five cases are on record of a de novo unbalanced translocation t(Y;1)(q12;q21), mostly seen in mosaic state; this translocation comprises a
majority of the total of eight known cases with an essentially complete 1q trisomy (Scheuerle et al., 2005). Presumably, similar sequences on 1q21 and Yq12 predispose to this recurring rearrangement. The phenotype, unsurprisingly, is very severe.
X-Y TRANSLOCATIONS The Classical X-Y Translocation Of the major types of X-Y translocation, the classical form is the most frequently seen (Fig. 6–16a, b). The X and Y breakpoints are constant, at the cytogenetic level, involving Xp22.3 in the distal X short arm, and Yq11/q12 in the proximal Y long arm (Bukvic et al., 2010). It is, certainly, readily recognized cytogenetically and has the karyotypic notation 46,X or Y,der(X),t(X;Y)(p22.3;q11). The important genotypic defect is deletion of the distal Xp segment, with the loss including PAR1. At the molecular level, there is variation in the amount of Xp deleted, and the phenotype depends in part at least upon which of the following distal Xp genes may be lost: ARSE (arylsulfatase E, for chondrodysplasia punctata), SHOX (short stature homeobox, for Leri-Weill dyschondrosteosis), STS (steroid sulfatase, for ichthyosis), KAL (Kallmann syndrome), MRX49 and NLGN4 (mental retardation), and OA1 (ocular albinism). The Y-originating segment does not contain SRY. The (female) person who is 46,X,der(X)t(X;Y) has a partial monosomy for this Xp segment, and the (male) individual with 46,Y,der(X)t(X;Y) is partially nullisomic. The female t(X;Y) heterozygote is characteristically fertile and of normal intelligence. If SHOX is deleted, the monosomic state for this gene determines a particular form of short stature and wrist deformity (Leri-Weill dyschondrosteosis) (Calabrese et al., 1999; Bukvic et al., 2010). The male t(X;Y) hemizygote is typically the son of a t(X;Y) mother (Hsu, 1994). Some may be cognitively normal, in those in whom the breakpoint is more distal. Should a more proximal molecular breakpoint expose a mendelian “brain gene,” such as MRX49 and NLGN4 mentioned earlier, mental impairment results, and apparently this is more usually the case. If the male has Leri-Weill dyschondrosteosis, it is no more marked than in the female, reflecting the fact that the SHOX locus is in the PAR1, and each sex still retains one copy of the gene, on their normal X or Y, respectively. Infertility is almost invariable, Sex Chromosome Translocations •
133
FIGURE 6–16 Four more frequent ways in which X-Y translocations are seen. (a) The classical t(X;Y) (p22.3;q11) together with a normal X (in a female). (b) The classical t(X;Y) together with a normal Y (in a male). (c) The cryptic t(Xp;Yp), with the Yp segment containing the SRY gene, in a “46,XX male.” (d) The cryptic t(Xp;Yp) as the sole gonosome, in a “45,X male.” Y chromatin: white indicates Y euchromatin, black indicates that part of distal Yp euchromatin encompassing the pseudoautosomal region and the SRY locus, and crosshatching indicates Yq heterochromatin. Note that gonadal sex accords with the absence (a) or presence (b–d) of the SRY gene.
due to spermatogenic arrest (Gabriel-Robez et al., 1990). Sperm production has, however, been documented in one case, albeit at a very low level (125,000 per ml), in a man with the typical 46,Y,der(X),t(X;Y)(p22.3;q11) karyotype (Morel et al., 2001). He was of normal intelligence, height 165 cm (5 feet 5 inches), with normal external genitalia and normal endocrine indices, and he had presented with infertility. There were equal numbers of 23,der(X) and normal 23,Y sperm, but about 20% of sperm were otherwise abnormal, the most common defect being 24,Y,der(X). The majority of cases are familial. Presumably, the X-Y chromosome arose following a reciprocal exchange between the X and Y during spermatogenesis in the individual fathering the originating (female) translocation carrier in the family. This event is facilitated by the apposition of X and Y segments having a high degree of homology; for example, a crossover between the Kallmann locus on the X chromosome and a Kallmann-like nonfunctional pseudogene on the Y chromosome long arm (Guioli et al., 1992). In the female, the pattern of X-inactivation tends toward inactivation of the der(X)t(X;Y), but this is variable and unpredictable (Gabriel-Robez et al., 1990). 134
•
Some X-Y translocations cytogenetically apparently identical to the classical type are associated with the rare syndrome of microöphthalmia with linear skin defects (MLS) (Kotzot et al., 2002). The phenotype results from aberrant expression of the HCCS gene at Xp22, which encodes a synthase producing cytochrome c, better known as one of the components of the mitochondrial respiratory chain, but which also has a role in inducing apoptosis (Wimplinger et al., 2006). Disordered apoptosis may be the basis of the tissue defects in MLS. T HE CR YPT I C Xp -Yp T R A N S LO CAT I O N ( “XX M A L E” A N D “ 4 5 , X M A L E” )
This form of the X-Y translocation is usually not visible (or barely visible) to the cytogeneticist without the use of fluorescence in situ hybridization (FISH) using Yp sequences as probe. Again, the X breakpoint is within Xp22.3; but the Y breakpoint is in the short arm, proximal to the testis-determining gene (SRY). The genotypic consequences are loss of the distal region of the X chromosome and the transfer of the SRY gene onto an almost intact X chromosome. Thus, the person is male. The karyotype would initially appear to the cytogeneticist either as 46,XX or as 45,X (Fig. 6–16c, d). In truth,
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
it is 46,X,der(X)t(X;Y)(p22.3;p11) or 45,der(X) t(X;Y)(p22.3;p11). This translocation accounts for most supposed XX males and some 45,X males. If there is loss of one copy of the SHOX gene, LeriWeill dyschondrosteosis (see earlier) is the expected consequence (Stuppia et al., 1999). The MLS syndrome, mentioned earlier, has been observed (Anguiano et al., 2003). The translocation arises from an abnormal X-Y recombination during paternal meiosis (Weil et al., 1994). Almost always, it occurs sporadically, and the affected males are infertile, although an extraordinary familial exception is recorded in Sharp et al. (2004). Two individuals, likely distantly related, presented with ovotesticular disorder of sex development. It may be that SRY in these two was partially operating, due to a variably penetrant position effect. “ 46, X,d e l (Yq )” W ITH CRY PT I C Xq-Yq I NT E R C H A N G E
A third category is the X-Y translocation arising de novo from an exchange during paternal spermatogenesis between Yq and distal Xq, producing an apparent del(Yq) chromosome that actually contains a very small segment of distal Xq (Lahn et al., 1994). The karyotype could be written 46,X,der(Y) t(X;Y)(q28;q11.2). The functional distal Xq disomy produces a characteristic severe phenotype (Sanlaville et al., 2009). In the female, the reciprocal 46,X,der(X) karyotype might hypothetically imply a mild phenotype, essentially reflecting a very small distal del(Xq), and assuming that the abnormal X would be preferentially inactivated. Cheng et al. (2009) provide a rare example of an Xq;Yq female, a 33-year-old woman who presented with premature ovarian failure, and whose karyotype they determined to be 46,X,der(X)t(X;Y)(q26.3;q11.223). The X chromosome could be appreciated as subtly abnormal on classical cytogenetics; it took comparative genomic hybridization (CGH) to discover the Yq material, and then FISH to reveal the true nature of the rearrangement.
Other Variant Forms Other forms of t(X;Y), typically but not always arising de novo, are often associated with infertility, and in some with intellectual deficit. The possibilities are listed and illustrated in Hsu (1994) and include der(X) chromosomes from translocations of varying lengths of Yq to a breakpoint at various levels on
Xp or Xq, and der(Y) chromosomes from translocations of varying small lengths of Xp to a breakpoint at various levels on Yq. The dicentric X-Y translocation comprises an almost complete Y attached at a distal Yp breakpoint to an X chromosome, at either an Xp or an Xq breakpoint. For example, Baralle et al. (2000) describe a girl with a Yp to Xp rearrangement, the karyotype 46,X,dic(X;Y)(p22.3;p11.2), who presented with Leri-Weill dyschondrosteosis. Her pubertal development was regarded as being normal, although she had yet (at age 14 years) to undergo menarche. Her femaleness was due to absence of the SRY gene, the breakpoint on Yp being proximal to its locus. A girl who presented at age 10 with an ovarian cancer, and in whom the X breakpoint was in the proximal long arm, her karyotype 46,X,der(Y)t(X;Y)(q13.1;q11.223), is reported in Lissoni et al. (2009); her female gender may have reflected inactivation of SRY from the influence of the XIC upon the der(Y).
X-X TRANSLOCATIONS The general karyotype is written 46,X,der(X), t(Xp;Xq), and the resultant imbalance is a dup/del of Xp/Xq, or vice versa. The translocation could have arisen following unequal recombination between the two X chromosomes in the oöcyte. Or the rearrangement could have occurred within the one X chromosome, folding in upon itself, in which case the origin is more likely paternal (Giglio et al., 2000). Xp11.23 and Xq21.3 are favored as breakpoints, and the translocated del/dup segments may therefore be large. Pubertal and/or menstrual abnormality is the usual presentation, and infertility is the rule (Letterie, 1995). Maternal transmission is, however, recorded in Reinehr et al. (2001), concerning a mother and daughter with short stature both having a t(X;X) (p22;q26) chromosome. These breakpoints are distal, and thus the del/dup segments in this case are small. The monosomic Xp segment included the SHOX gene, and this presumably was the cause of the short stature. The mother had had a normal menstrual history, and she had two other healthy children normal heights.
Y-Y TRANSLOCATIONS For the sake of completeness, the presumed existence of the very rare Y-Y translocation is noted. Some may in fact have been isodicentric Yq chromosomes (Hsu, 1994; Hernando et al., 2002). Sex Chromosome Translocations •
135
GENETIC COUNSELING THE X-AUTOSOME TRANSLOCATION Fertility is affected in the X-autosome heterozygote and hemizygote. Approximately half of the female carriers, and practically all males, are likely to be infertile.
modified by spreading of inactivation, but this is unpredictable. (4) Any 2:2 unbalanced segregant from a doublesegment translocation (Fig. 6–7h– j) has a combined duplication/deficiency, and spontaneous abortion is probable. But spreading of inactivation in a female conception may attenuate a partial autosomal trisomy and allow for survival, albeit with phenotypic defect. (5) Adjacent-2 possibilities need individual assessment (Fig. 6–8).
The Female Heterozygote If fertile, the female heterozygote has a substantial risk for having abnormal offspring due to an imbalanced chromosomal constitution. At one end of the scale, the abnormality might be mild (e.g., partial Klinefelter syndrome) or barely discernible (e.g., partial X trisomy). At the other end, it could be severe (e.g., partial X disomy or autosomal aneuploidy, not modified by inactivation). The counselor should determine the theoretical gametic combinations from the particular category of translocation, with reference to the examples described in the section on “Biology.” Adjacent-1 and 3:1 tertiary trisomy are the major malsegregation modes to be considered. Figures 6–7 and 6–8 provide a guide; but each translocation needs to be assessed on its own merits. General comments follow. (1) A single-segment translocation with an X segment of large size would imply risks for partial Turner, partial Klinefelter, and partial XXX syndromes (Figs. 6–2 and 6–4; Fig. 6–7b, c). (2) A single-segment translocation with an X segment of small size would imply a risk not only for one of these three partial gonosomal aneuploidies but also for functional disomy or nullisomy for a small distal Xp or Xq segment. A functional disomy would have a severe outcome (Fig. 6–5b, segregation as per heavy arrows; Fig. 6–7a). Nullisomy in the male, for all but the smallest segments, would be lethal in utero (Fig. 6–5b, segregation as per light arrows; Fig. 6–7b). The borderline between viable (but severe) and nonviable Xp deletion in the male may be at Xp22.2, in which about 10 Mb of DNA is removed (Melichar et al., 2007). (3) A single-segment translocation with an autosomal translocated segment of “viable size” (Fig. 6–7d–f ) implies a risk for partial autosomal monosomy or trisomy from adjacent-1 segregations. In the female conceptus, the trisomy may be 136
•
T HE L EV EL O F R I S K
The risk for many female heterozygotes, who are fertile, will be “substantial.” An otherwise nonviable unbalanced conception may survive because inactivation tempers the imbalance; and some conceptions with the structurally balanced complement may be functionally unbalanced due to aberrant inactivation patterns. The counselor should go through the exercise of determining possible malsegregant outcomes, as depicted in Figures 6–7 and 6–8. The risks to have a liveborn child with a structural and/or functional aneuploidy may be in the range 20% or higher. In Stene and Stengel-Rutkowski (1988), and with specific reference to singlesegmental translocations involving Xp, the risk for adjacent-1 malsegregants was 24%, although interestingly the risk associated with 3:1 segregation leading to interchange trisomy X was very low, less than 0.8%. As we discussed earlier, the components making up the total risk may comprise a very mild abnormality through to severe mental and physical defect. There is the difficulty of knowing what risk might apply to a child with the same balanced translocation (see later discussion). Only with the 46,XX and 46,XY karyotype can one be confident of normality, other things being equal. Even more than with the common autosomal translocation, the risks relating to each X-autosome translocation will be specific to that particular rearrangement, and extrapolation from other translocations will be fraught. Panasiuk et al. (2004) have made a start, in deriving specific risk figures for four different translocations, in each case the X breakpoint involving the short arm (Table 6–2). The one circumstance in which they consider data pooling to be permissible is in rearrangements in which the autosomal breakpoint is in the short arm of an acrocentric.
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Table 6–2. Estimated Risk Figures for Having a Liveborn Aneuploid Child, or a Child Stillborn or Dying as a Neonatea, Because of Imbalance due to X-Autosome Malsegregation (Adjacent and/or 3:1), in Four Specific Translocations, Three Double Segment, and One, the X;22, Effectively Single Segmentb
TRANSLOCATIONS
RISKC (%)
t(X;5)(p22.2;q32) t(X;6)(p11.2;q21) t(X;7)(p22.2;p11.1) t(X;22)(p22.1;p11.1) e
4.2 3.3d 2.1 17
a Figures may be considered as expressing the percentage risk to have an aneuploid liveborn or stillborn infant, from a pregnancy which had proceeded to at least 28 weeks gestation. b Families published or cited in Panasiuk et al. (2004). c The figure in one family (X;22), in which the autosomal breakpoint is in the p arm of an acrocentric chromosome, comes from direct segregation analysis, and combining with literature cases of another X;acrocentric p arm translocation (of chromosome 15). In the remaining three, the figure is indirect and derived from applying this rule: halving the risk for the lesser of the two risks, which would otherwise have applied to each translocated segment when viewed as a single-segment imbalance. d This carrier mother had presented having had an unkaryotyped malformed stillbirth at 42 weeks gestation, suggesting that at least one of the malsegregant combinations might be compatible with survival to term. The figure of 3.3% might thus be an underestimate, if the risk figure were taken to include stillbirth. e This translocation is very close to, and might possibly be the same as, the t(X;22) in Figure 6–11.
The Male Hemizygote Infertility is almost inevitable, barring the possibility of medical intervention (Ma et al., 2003). If sperm can be accessed, from an ejaculate or via testicular sperm extraction, in vitro fertilization (IVF) using ICSI may be attempted; greater spermatogenic success may attend translocations with pericentromeric or centromeric breakpoints (Perrin et al., 2008). The outcomes from which normality could be expected, other things being equal, are the two alternate segregations leading to 46,XY, and to 46,X,t(X;A), respectively; the likelihood for this may be about 50% (as extrapolated from sperm data in one case; Perrin et al., 2008). Normality in a (necessarily heterozygous) daughter would require
inactivation to have been skewed in favor of the normal X; the likelihood for this to have happened may be greater in the case of larger translocated segments. Phenotypic normality in the hemizygous father would allow the inference of a truly balanced rearrangement, and thus no question should arise about a cryptic deletion/duplication. The presumption of an approximately 50% risk was, for example, brought to the attention of the man with an (X;18)(q11;p11.1) translocation noted earlier (see “Biology,” and Fig. 6–12); nevertheless, and given the practical matter of a long wait for PGD, the couple went ahead with IVF and ICSI, having prenatal diagnosis in the pregnancy, and they had a normal 46,XY son (Perrin et al., 2008). Ma et al. (2003) describe a successful outcome in a man with a whole-arm translocation, t(X;20)(q10;q10), maternally inherited, and from whose ejaculate only about 50 sperm were able to be retrieved; an embryo created at IVF went on to become a carrier daughter, who was normal physically and developmentally on assessment at age 12 months. The child, and her heterozygous paternal grandmother, both displayed skewed X-inactivation. Interestingly, the man’s brother, also an X;20 hemizygote, had had 7 years of infertility, but then had two normal daughters. These girls’ karyotypes, and paternity status, had not been evaluated. The Balanced Inherited X-Autosome Detected Prenatally. This can be a vexed question. Consider the circumstance of a phenotypically normal carrier mother, who has prenatal diagnosis, or PGD. A balanced X-autosome karyotype identified in a female fetus, or in a female embryo (the balanced translocation, and normal states not routinely distinguishable), might well eventually lead a normal daughter, but this cannot be made as a firm statement. As yet, we lack a good understanding of the relative roles of aberrant X-inactivation, versus gene disruption at a breakpoint, in causing phenotypic abnormality. If it were the former, then a normal carrier mother (and she having a “perfect” 100:0 normal: derivative X-activation ratio) could have an abnormal carrier daughter, if the daughter’s inactivation ratio were “imperfect.” The case in Figure 6–9 might exemplify this scenario (or might not, if the child’s abnormality had been coincidental). But if the latter, then the mother’s normality would, of itself, indicate absence of gene disruption; and presumably her daughter would inherit the same intact translocation, and herself be normal, other things being equal. There is a Sex Chromosome Translocations •
137
need for a large study, with unbiased ascertainment, of the carrier daughter offspring of the (normal) carrier mother, in order to address this issue. A most remarkable scenario is that of “incorrect” inactivation of the der(X) comprising the “first hit” in a tumor cascade. A mentally handicapped woman in her twenties developed schwannomas, and she was found to carry a de novo t(X;22)(p21.3;q11.21); these breakpoints are very close to the t(X;22) discussed elsewhere and shown in Figure 6–11. Although the X-inactivation pattern on blood was appropriate, in tumor tissue the der(X) was inactivated (Bovie et al., 2003). This inactivation may have spread through to the 22q-segment, which contains two loci associated with schwannoma susceptibility (the NF2 and SMARCB1 loci). Too little information exists concerning the phenotype of the male hemizygote born to a female X-autosome heterozygote for any firm advice to be offered. Normality has been recorded in this setting, but so has major genital defect, which in one case was the consequence of compromised function of the androgen receptor gene (Buckton et al., 1981; Kleczkowska et al., 1985; Callen and Sutherland, 1986; Ma et al., 2003). Fetal ultrasonography may be useful to check for normal male genital development. This approach was offered to the mother whose karyotype appears in Figure 6–11, and who had a 46,Y,t(X;22) result at amniocentesis in her second pregnancy. A normal baby boy was subsequently born, whose infant development was quite normal. Otherwise normal male carriers would almost certainly be infertile (see earlier).
Y-AUTOSOME TRANSLOCATIONS The Apparently Balanced Y-Autosome Translocation It is notable that the same balanced Y-autosome translocation can behave differently in different male members of a family in terms of fertility, this presumably reflecting the importance of the background genetic contribution to the control of the mechanics of spermatogenesis (Teyssier et al., 1993; Rumpler, 2001). For those who are fertile, risk data are too few to form a secure base for genetic counseling. From first principles, unbalanced forms are probable, several of which will often be viable (according to the autosome in question, and the site of the autosomal breakpoint), and the option of
138
•
prenatal diagnosis is appropriately offered or, and especially if IVF is needed, PGD. As discussed in the “Biology” section, in spite of there being several more imbalanced than balanced possibilities, there are tentative grounds for supposing that alternate segregations (normal and balanced forms) may be favored. The t(Y;8) family of Sklower Brooks et al. (1998) noted earlier and shown in Figure 6–13 demonstrated three of the four predicted alternate and “adjacent-1” karyotypic outcomes: 46,XX, the 46,X,t(Y;8) balanced carrier, and 46,X,der(Y), the former two outnumbering the latter. The 46,X,der(Y) karyotype produced sons with an 8p duplication; the other unbalanced karyotype, 46,XX,der(8), would have produced a daughter with an 8p deletion. Manifestly, the carrier male, while he could have a normal daughter, could never conceive a 46,XY child. Sperm karyotyping, if available, may be a helpful investigation. In the man with the rare 13p;Yp fusion mentioned earlier (Alves et al., 2002), having demonstrated that most sperm had a balanced complement, reassurance could be offered, in this particular case, that if pregnancy were achievable there would be a good chance of producing a normal child. For the infertile man, assisted reproduction may offer the possibility of paternity. A sperm count way below the level needed for natural conception may yet allow retrieval of sperm for ICSI. Testicular aspiration may provide sperm even when they are completely absent in the ejaculate. With the need for IVF, PGD becomes attractive because of the probable substantial genetic risk, in most cases, for unbalanced forms, and considering the practical point that the embryo is nicely accessible. Taking the example of the oligospermic man with a 46,X,t(Y;18) (q11.2;q21) karyotype, shown in Figure 6–14, he could, in theory, and through IVF, have a 46,XX daughter, and a 46,X,t(Y;18) son like himself. The substantial fraction of unbalanced forms that could be viable in this case, out of the 16 total possible conceptions, does becomes a relevant matter at PGD. These issues of IVF and PGD are discussed in more detail in Chapter 26. THE Yqh-ACROCENTRIC TRANSLOCATION
Probably, these translocations can be regarded as being no more than interesting variant chromosomes, and of no clinical significance. In the case of the t(Yq;15p), a theoretical risk for trisomy 15 with
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
correction to UPD (White et al., 1998; RajcanSeparovic et al., 2001) is neither to be completely ignored nor to be overstated. The “45,X” Yp-Acrocentric Translocation. These chromosomes are probably stable, and not (if fertility is achievable) implying a risk for phenotypically abnormal offspring (Callen et al., 1987).
THE CLASSICAL X-Y TRANSLOCATION The female with an X-Y translocation is usually fertile and of normal intelligence. She has a 50% risk for having a child, whether a son or daughter, who would have the translocation. An X-Y translocation son may be abnormal, according to the extent of distal Xp nullisomy and the loci involved (Seidel et al., 2001). If the mother is short, an X-Y translocation daughter would also be short, likely because of deletion of the SHOX locus. As with Turner syndrome, growth hormone treatment may be appropriate for such a child. She would probably be, like her mother, fertile. A child receiving the mother’s normal X would of course be normal, 46,XX or 46,XY. Prenatal diagnosis is appropriately offered. The male X-Y translocation carrier is almost invariably infertile. A sperm chromosome study has been undertaken in only one der(X)t(X;Y) man,
referred to in the “Biology” section earlier (Morel et al., 2001). He had severe oligozoöspermia, and notably sex chromosome aneuploidy was recorded in 20% of sperm. Otherwise, 40% of sperm were normal 23,Y, and 40% had the t(X;Y). Conception in such a case could only ever be achieved via IVF. If preimplantation diagnosis were to be attempted, the choice of a 46,XY embryo (the only normal gonosomal possibility) would allow avoiding the genetic risk for the next generation.
X-X TRANSLOCATIONS Infertility is the expectation, and a theoretical question of genetic risk will usually be academic. In a small imbalance, fertility may be retained, as in the example of Reinehr et al. (2001) discussed earlier. The genetic risk would, in essence, be the same as for the 46,X,abn(X) heterozygote. A daughter receiving the X-X translocation would be expected to have a phenotype similar to that of her mother. A male pregnancy would be very likely to miscarry at any early stage, due to an X nullisomy/disomy. If the del/dup segments were very small, viability might be possible, but with probable major phenotypic defect. Children receiving the mother’s normal X chromosome would of course be normal, other things being equal.
Sex Chromosome Translocations •
139
7 robertsonian translocations
THE AMERICAN INSECT CYTOGENETICIST W. R. B. Robertson first described translocations of chromosomes resulting from the fusion of two acrocentrics in his study of insect speciation in 1916, and this type of translocation is named Robertsonian (abbreviation rob) in his honor. There are five human acrocentric autosomes—chromosomes 13, 14, 15, 21, and 22 (the 13, 14, and 15 are the D group chromosomes, and the 21 and 22 comprise the G group)—and all are capable of participating in this type of translocation. The composite chromosome produced includes the complete long arm chromatin of the two fusing chromosomes, although it lacks at least some of the short arm chromatin. Robertsonian translocations are among the most common balanced structural rearrangements seen in the general population with a frequency in newborn surveys of about 1 in 1000 (Blouin et al., 1994). Historically, the most important Robertsonian translocations are the D;21 and G;21, which are the basis of most familial translocation Down syndrome. Uniparental disomy is of relevance, with respect to the two imprintable acrocentrics, chromosomes 14 and 15. 140
•
In this chapter, we consider the case of the phenotypically normal person who carries, in balanced form, a Robertsonian translocation. We generally use a short cytogenetic description for the carrier state, thus, 45,XX,rob(14q21q) or simply rob(14q21q). The formally correct ICSN designation for a short arm to short arm fusion Robertsonian translocation is, for example, 45,XX,der(14;21)(q10;q10) or 45,XX,rob(14;21)(q10;q10).
BIOLOGY The great majority of balanced Robertsonian translocations involve two different chromosomes (a heterologous or nonhomologous translocation); those involving the fusion of homologs (homologous translocation) are very rare. Heterologous translocations can be transmitted through many generations of phenotypically normal heterozygotes, whereas the homologous translocation is almost always seen as a de novo event in the consultand. As Table 7–1 attests, the rob(13q14q) and the rob(14q21q) are predominant. If we exclude the rob(21q21q)—most
1
2
3
6
7
8
13
14
15
19
20
9
4
5
10
11
12
16
17
18
21
22
X Y
14
FIGURE 7–1
The balanced rob(13q14q) in a phenotypically normal male.
1
2
3
6
7
8
9
4
5
10
11
12
16
17
18
21
22
X Y
21
FIGURE 7–2
13
14
19
20
15
The balanced rob(14q21q) in a phenotypically normal male.
FIGURE 7–3
1
2
3
6
7
8
13
14
15
19
20
TRANSLOCATION LITERATURE UNBIASED REVIEW ASCERTAINMENT
3% 33% 2% 2% 1% ½% 2% 30% 1% 2% 3% ½% 17% 2% 1%
2% 74% 2% 1% 2% – 5% 8% 2% – ½% 1% 3% ½% –
Note: Relative frequencies in literature review (most cases being of biased ascertainment), and in studies in which ascertainment was unbiased. *Most are i(21q) Down syndrome; the figure for true rob(21q21q) is probably nearer ½%. Source: From Hook and Cross (1987b) and Therman et al. (1989).
142
•
5
10
11
12
16
17
18
21
22
X
The unbalanced rob(14q21q) in a girl with translocation Down syndrome.
Table 7–1. The Frequency of Robertsonian Translocations
13q13q 13q14q 13q15q 13q21q 13q22q 14q14q 14q15q 14q21q 14q22q 15q15q 15q21q 15q22q 21q21q* 21q22q 22q22q
9
4
of which are actually isochromosomes for 21q—the rob(13q14q) accounts for around 75% of all Robertsonian translocations in unbiased studies, and indeed it is the commonest single chromosome rearrangement in the human race. Since 1 in 1000 persons is a rob heterozygote, the prevalence of the rob(13q14q) carrier is about 1 in 1300. Karyotypes of the 13q14q and 14q21q carrier states, and of the unbalanced 14q21q state leading to translocation Down syndrome, are shown in Figures 7–1 through 7–3. Balanced carriers for any of the five homologous translocations seem to be of about equal rarity.
Formation of the Translocation There are three possible mechanisms of formation of the balanced heterologous translocation: fusion at the centromere (centric fusion), union following breakage in one short arm and one long arm (essentially, a whole-arm reciprocal translocation), and union following breaks in both short arms (Guichaoua et al., 1986) (Fig. 7–4). The first two mechanisms are rare (if ever, in the case of centric fusion) and would produce a translocation chromosome with one centromere (monocentric), while
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(b)
(a)
or
(c) FIGURE 7–4 Mechanisms of formation of Robertsonian translocations. (a) Centric fusion, giving a monocentric chromosome; (b) breakage in one short arm and one long arm, giving a monocentric; and (c) breakage in both short arms, giving a dicentric or, after suppression of one centromere, a monocentric.
the third results in a chromosome with two centromeres (dicentric). The common rob(13q14q) and rob(14q21q) translocations are practically always dicentric and are formed predominantly during female meiosis, with consistent breakpoints at the molecular level (Bandyopadhyay et al., 2002). In some dicentrics, one centromere is “suppressed,” and the chromosome appears monocentric. This heterogeneity of formation is not of any clinical significance that can presently be discerned. In the reciprocal type, the other product may rarely survive as a stable small bisatellited marker (Schmutz and Pinno, 1986). The propensity to recombine may be the consequence of recombination between similar sequences shared by acrocentric chromosomes. The predominance of the rob(13q14q) and the rob(14q21q) may be due to specific homologous but inverted segments in these pairs of chromosomes that encourage crossover, while the variable breakpoints in the uncommon translocations point to a more random process (Bandyopadhyay et al., 2001a, 2001b, 2002). Rare cases are due to postzygotic joining together, a point that can be proven when the two component chromosomes can be shown to have come one from one parent, and one from the other
(Bandyopadhyay et al., 2003). Just as a Robertsonian translocation can form de novo from the fusion of chromosomes, so can it (very rarely) revert to two separate chromosomes by a “back-mutational” fission (Pflueger et al., 1991) (and see p. 159). The balanced homologous Robertsonian chromosome may arise from fusion in the zygote of the paternal and maternal homologs, in which case it is a true translocation. The site of formation may be at the first mitosis, a conclusion we drew from studying a woman with 45,XX,rob(13q13q), who showed no mosaicism on biopsy samples from a number of different tissues taken during surgery for tubal ligation (Gardner et al., 1974). Alternatively, it may be an isochromosome, with the stage having been set in meiosis: a nullisomic egg due to a maternal nondisjunction leads to a monosomic conceptus, which is then “rescued” by reduplication of the paternal homolog as an isochromosome, and thus with uniparental disomy for the chromosome in question (discussed later). Berend et al. (1999) showed a de novo 45,i(13q),upd(13)pat in a normal infant to have complete isozygosity for chromosome 13 markers, indicative of this scenario of postzygotic monosomy rescue. In another instance, they could show a paternal meiotic origin of the i(13q) in a normal adolescent with 45,i(13q),upd(13)pat. This individual would have had trisomy 13, had it not been for gametic complementation: the mother contributed a nullisomic 13 ovum (she being, by extraordinary coincidence, a 13q14q heterozygote). These two cases came to attention only through fortuitous discovery at prenatal diagnosis. Nucleolar Organizing Regions and the Robertsonian Translocation. The nucleolar organizing regions (NORs) are located in the “stalks” of the short arms of the acrocentric chromosomes, in the p12 regions, and comprise multiple copies of genes coding for ribosomal RNA. Not all NORs are active: as judged by silver (Ag-NOR) staining, most individuals have four to seven per cell that are functioning (Varley, 1977). Presumably, there is a minimum requirement for normal cellular function. When a Robertsonian translocation forms, the NORs of two of the fusing chromosomes are lost, at least with the rob(13q14q) and rob(14q21q). Thus, an individual with a Robertsonian translocation has only eight acrocentric short arms and therefore eight NORs, but this is nevertheless a sufficient number. Robertsonian Translocations
•
143
THE HETEROLOGOUS ROBERTSONIAN TRANSLOCATION D E TAILS O F MEIOTI C BE HAV I OR
This type of Robertsonian translocation chromosome comprises the long arm elements of two different acrocentric chromosomes. At meiosis in the heterozygote, the translocation chromosome and the two normal acrocentric homologs synapse as a trivalent. Following 2:1 segregation, six types of gamete are produced (Fig. 7–5). “Alternate” segregation leads to the production of normal and balanced gametes; and adjacent segregation produces two types of disomic and two types of nullisomic gamete. 3:0 segregation occurs, but it is rare. In obvious contrast to what happens with the reciprocal translocation, the chromosomally abnormal conceptuses have a complete aneuploidy. Only unbalanced conceptuses that are effectively trisomic for chromosome 13 or 21 can survive substantially through the course of the pregnancy (whether to fetal death in utero, stillbirth, or live birth). Fetal trisomy 14, 15, and 22 are expected to end in miscarriage in the first or early second trimester; and any of
the monosomies would abort in the very early first trimester, possibly before the time of implantation. Of these six possible outcomes (eight if we include 3:0 segregants), some are more likely to occur than others. Judging from the outcomes at birth, one might conclude that alternate segregation is favored. From the male heterozygote, translocation Down syndrome (DS) and translocation trisomy 13 are scarcely ever seen in the offspring, and in a fairly small minority for DS, 10%–15%, from the female (Table 7–2). But of course, as just mentioned, there has been the complete selection in utero against some unbalanced forms, and a variable selection against the two potentially viable imbalances, trisomy 13 and trisomy 21. Segregation at the level of the gamete is a different story. Sperm and oöcyte studies show considerable fractions of unbalanced forms. (Naturally, most if not all of the individuals proceeding to gamete testing in these reported studies will have experienced reproductive difficulty, and thus the data from their gametes may not necessarily be applicable to the larger number of carriers with apparently normal fertility.) Ogur et al. (2006) have reviewed the literature on sperm analysis and contributed their own
FIGURE 7–5 Meiotic behavior of the Robertsonian translocation. (a) Trivalent at synapsis. (b) Normal and (c) carrier gametes from “alternate” segregation. (d) Disomic and (e) nullisomic gametes from adjacent segregation. Note that there are six possible combinations (ignoring 3:0 segregation), of which two are normal/ balanced, and four are unbalanced. 144
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Table 7–2. Estimates of Risks to Have a Child with Aneuploidy or with a Uniparental Disomy Syndrome, for the Heterologous rob Carrier CARRIER PARENT MOTHER
FATHER
rob
UNBAL.
UPD*
UNBAL.
UPD*
13q14q 13q15q 13q21q 13q22q 14q15q 14q21q 14q22q 15q21q 15q22q 21q22q
1% 1% 10%–15% 1% – 10%–15% – 10%–15% – 10%–15%
<½% <½% – – ½% <½% <½% <½% <½% –
<1% <1% <1% <1% – <1% – <1% – <1%
<½% <½% – – <½% <½% <½% <½% <½% –
Note: Estimates for the uncommon rob translocations are extrapolated from data for the common robs. Unbal., unbalanced, with a full aneuploidy for chromosome 13 or 21; UPD, uniparental disomy; UPD*, abnormal child with syndrome of UPD 14 or UPD 15.
cases, and their findings are set out in Table 7–3. Across all types of translocation (and it is likely that no real differences exist), 81%–92% of sperm are normal/balanced due to alternate segregation, and 7%–19% unbalanced due to adjacent segregation. Rounding these figures, 10%–20% of sperm are unbalanced, and 80%–90% are balanced. On oöcyte analysis, using the ingenious approach of fluorescence in situ hybridization (FISH) analysis on polar bodies, Munné et al. (2000a) determined in four 45,XX,rob(14q21q) carriers an average 42% of unbalanced forms, and seven 45,XX,rob(13q14q) carriers with an average 32%; others have provided similar data (Table 7–3), and Bint et al (2011) assemble a recent review (and see p. 437). In the important 14q21q group, about 20% of ova may be disomic 21, and evidently half or more of these are able survive through to term, to give the 10%–15% risk figure for a child with DS, mentioned earlier. Meiotic Drive. Meiotic drive is an influence whereby one of the products at meiosis may be favored and have a better than even chance of coming to be in the successful gamete. The Robertsonian translocation provides an apparent example. At the level of the offspring produced, de Villena and Sapienza (2001) demonstrated that children of
female carriers of rob translocations have a ratio close to 60:40 for the balanced rob compared to normal karyotypes. No such effect could be confirmed for the male rob carrier. Daniel (2002) has confirmed these interpretations in a retrospective analysis of prenatal diagnosis data, with rigorous attention to the need to avoid bias, showing a 116:81 ratio in favor of balanced carrier offspring compared to normal karyotypes where the mother is the carrier parent, compared to a 42:41 ratio for carrier fathers. At the level of the gamete, Ogur et al. (2006) showed that, among the unbalanced forms in sperm, nullisomies outnumber disomies. POST ZYG OT I C “ CO R R ECT I O N ” A N D A SS O CI AT ED UN I PA R EN TA L D I S O MY
Trisomic Correction. An initial translocation trisomy may be “corrected” by mitotic loss of one of the free homologs and lead to uniparental disomy (UPD) in the embryo. For example, a presumed mechanism whereby UPD 15 could arise from a rob(13q15q) parent is outlined in Figure 7–6. Essentially, adjacent segregation produces a trisomic 15 conception, and then loss of the chromosome 15 contributed from the other parent,1 at an early Robertsonian Translocations
•
145
Table 7–3. Segregation Patterns in Gamete Studies upon Heterologous Robertsonian Heterozygotes (Several of Whom Had Presented with Infertility) TRANSLOCATION
SEGREGATION PATTERN
SPERM
2:1 ALTERNATE
2:1 ADJACENT (fractions with the viable disomy)
t(13q;14q) t(13q;15q) t(13q;21q) t(13q;22q) t(14q;15q) t(14q;21q) t(14q;22q) t(15q;22q) t(21q;22q) Range of the means
74%–92% (85%) 76%–93% (82%) 87%–88% (88%) 86% 91%–93% (92%) 72%–93% (87%) 79%–81% (80%) 90% 60%–97% (81%) 81%–92%
8%–26% (14%) 7%–23% (17%) 11%–12% (11%) 14% 7%–8% (7%) 7%–13% (10%) 19%–20% (19%) 10% 3%–40% (19%) 7%–19%
OÖCYTES
2:1 ALTERNATE
2:1 ADJACENT (fractions with the viable disomy)
68% 26% 60% 40% 58% 43% 40%–68%
32% (10%) 68% (16%) 20%* 50%* 42% (20%) 57% (0%) 32%–68%
a
t(13q;14q) t(13q;14q)b t(13q;14q)c t(13q;14q)c t(14q;21q)a t(14q;21q)b Range
Notes: The numbers of male subjects range from 25 (the common 13;14), to one per translocation type (the 13;22 and 15;22 cases). Each female subject is listed individually, six in all. Just over 24,000 sperm were studied, but only 200 analyzable eggs. Only 2:1 segregants are listed: 3:0 segregants are very rare, in sperm at least (three 3:0 egg segregants are footnoted below*). 2:1 alternate segregants would produce a normal or balanced karyotype in the conceptus; 2:1 adjacent segregants would produce trisomy or monosomy. Figures in parentheses are average ranges (sperm data), and fractions of gametes (oöcyte data), that could have produced the relevant viable aneuploidy, trisomy 13 or trisomy 21, accordingly. See also the review of Bint et al. (2011). Sperm data: From the review in Ogur et al. 2006, plus 13;15 and 14;22 cases from Moradkhani et al. 2006a and b, and with these other entries: 13;21 (Chen et al., 2007c; Hatakeyama et al., 2006), 13;22 (Anahory et al., 2005), 14;15 (Moradkhani et al., 2006a), 14;22 (Chen et al., 2005d), and 15;22 (Martin et al., 1992). Oöcyte (polar body) data: From aMunné et al. (2000a), bDurban et al. (2001), and cPujol et al. (2003). *Shortfall from 100% totals due to three 3:0 forms.
postzygotic stage, “corrects” the karyotype. UPD has no untoward effect if the chromosome is not subject to imprinting (except for the question of isozygosity for a recessive gene; see later), and chromosomes 13, 21, and 22 are in this category. If there is UPD for an imprintable chromosome—in this context, chromosome 14 or 15—a UPD syndrome would result. Engel and Antonarakis (2002) list just four recorded cases from a familial heterologous
rob: one of upd(14)mat, one upd(14)pat, and two of upd(15)mat. James et al. (1994) made a specific search in a group of 14 phenotypically abnormal rob carriers, in most of whom the translocation was inherited, and identified only one case of UPD. A similar retrospective study by Berend et al. (2002a) that included 30 phenotypically abnormal heterozygotes with a familial heterologous translocation revealed two patients with UPD, both having a
1 Note that with one or other chromosome 15 being the candidate to be lost, the risk for UPD to be generated is 50%. This is in contrast with correction in standard trisomy, in which, with three candidate chromosomes, the chances are 1 in 3 for the “wrong” one to be lost.
146
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
FIGURE 7–6 Uniparental disomy 15 from a rob(13q15q) parent, due to “trisomy rescue.” The heterozygous parent produces a malsegregant gamete with the translocation, and with a free chromosome 15. The conception thus has trisomy 15. Subsequently, as a postzygotic event, the chromosome 15 from the other parent is lost. Since most malsegregations will have been of maternal origin, the uniparental disomy (UPD) in this setting will usually be a maternal heterodisomy. (Chromosome 13 elements, white; chromosome 15 elements, crosshatched. The two chromosome 15 elements from the carrier parent are asterisked.)
rob(14q15q), one with upd(14)mat, and the other upd(15)pat. Thus, UPD due to a parental rob is extremely rare, with the worldwide total of reported cases barely numbering in double figures. The overall risk to have a child with a UPD syndrome, as determined from prenatal diagnosis data where one parent is a heterologous rob carrier, is 0.8% (see later). Residual Low-Level Trisomy. If the “correcting” mitosis occurs too late to include every cell that will contribute to the embryo proper, a translocation trisomic cell line may persist. The only example in the survey of Berend et al. (2000a) was that of a child with upd(13)mat from a rob(13q14q) mother, in whom a low level (4%) of trisomy 13 was shown on prenatal diagnosis, 45,XX,rob(13q14q)[48]/46, XX,+13,rob(13q14q)[2]. Bruyere et al. (2001) record in an abstract three such cases detected in a series of 281 prenatal diagnoses. Jenderny et al. (2010) showed 4%–8% translocation trisomy 13 mosaicism on blood, and 11% on buccal cells, in an abnormal boy whose karyotype could be written 45,XY,rob(13q14q)/46,XY,+13,rob(13q14q)mat; UPD was excluded. Monosomic Correction. Hypothetically, correction may also go the other way: that is, the conversion of a monosomic conceptus, due to a nullisomic gamete from 2:1 adjacent segregation in the rob parent, into a disomic conceptus. This conversion to disomy, or “correction,” would be achieved by the replication of the homolog contributed in the (normal) gamete of the other parent. A replicate free chromosome might be produced, in which case the karyotype would appear normal. Or the homolog could replicate as an isochromosome, which would produce the intriguing circumstance of a de novo Robertsonian-like chromosome in the setting of a true Robertsonian parent. This event, whichever one, would take place at a very early postzygotic stage and would necessarily lead to uniparental isodisomy (Berend et al., 2000a). It is, apparently, very rare and might usually fail unless it occurred by day 3, since monosomy becomes a lethal impediment after this stage (Ruggeri et al., 2004).
Association with Infertility There is an approximately 7-fold excess of Robertsonian heterozygotes among couples who are infertile, and a 13-fold excess among oligospermic 148
•
men (Tharapel et al., 1985; Ogur et al., 2006). A minority of rob carriers may have an individual predisposition, not necessarily shared by their heterozygous relatives, for a high frequency of unbalanced segregations, an insight that has been afforded by in vitro fertilization (IVF) studies. Alternatively, or additionally, the rob translocation may, of itself, compromise the fidelity of the first few mitoses, affecting mitotic segregation of the other chromosomes (Emiliani et al., 2003). By way of example, Conn et al. (1998) treated two couples with a Robertsonian translocation, who had been unable to achieve a normal pregnancy: a man with 45,rob (13q14q) and a woman with 45,rob(13q21q). They were able to karyotype a total of 33 day-3 embryos from the two couples. A considerable majority of embryos, almost 90%, were chromosomally abnormal. Of these, 40% were trisomic or monosomic for 13, 14, or 21 (some mosaic, and some double monosomic), and this might have been expected. Notably, 60% had a “chaotic karyotype,” in which the chromosome constitution varied randomly from cell to cell, and indeed the karyotype of the original zygote could not usually be determined.
M A L E I N F ERT I L I TY
As noted earlier, there is a 13-fold excess of rob heterozygotes among oligospermic men. A case has been made that, in this setting, synapsis is incomplete in the trivalent, and the heterochromatic regions of the short arms remain unpaired; these “exposed” regions then interfere with pairing in the X-Y bivalent, so that spermatogenesis is blocked from further progression (Luciani et al., 1987; Johannisson et al., 1987). Guichaoua et al. (1990) have directly observed the asynapsed short arms of the trivalent associating with the X-Y bivalent in testicular tissue from an oligospermic man heterozygous for a rob(14q22q), and Navarro et al. (1991) have similarly studied a rob(13q14q) man. Electron microscopic sperm analysis in a rob(14q22q) man with oligoasthenospermia showed marked ultrastructural defects in the great majority of spermatozoa, and attempted IVF was unsuccessful (Baccetti et al., 2002). Mice with several Robertsonian translocations show spermatogenic arrest if the translocations form a chain and associate with the sex chromosomes ( Johannisson and Winking, 1994). But it is notable that with some infertile 45,XY, rob(13;14) men, their brothers, fathers, or
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
other male relatives may have unimpaired fertility (Rosenmann et al., 1985). RARE CO MP LEXITIES
Robertsonian Fission. The Robertsonian translocations arise through a “fusion” of the short arm sequences. Equally, it can, in somatic tissues, revert to “normality” by fission (Perry et al., 2005). Although the resulting two acrocentric chromosomes would have somewhat truncated short arms that lacked NORs, this appears to be without any clinical consequence (and see also Chapter 8). Mosaicism for Two Robertsonian Translocations. A few examples are known of individuals with a 21-containing rob, such as 14q21q or 21q22q, plus an isochromosome of 21q, and presenting as a normal parent of a Down syndrome child, or with mosaic Down syndrome (Gross et al., 1996; Berend et al., 1998; Bandyopadhyay et al., 2003). Iwarsson et al. (2009) undertook sperm analysis in their patient with two Robertsonian translocations, a man whose karyotype of 45,XY, ro b ( 1 3 ; 1 3 ) (q 1 0 ; q 1 0 ) / 4 5 , X Y, ro b ( 1 3 ; 1 5 ) (q10;q10)dn had originally been identified in fetal life, at amniocentesis. Presenting some 22 years later for genetic counseling, a semen analysis revealed oligoasthenoteratozoöspermia, and on FISH study about 40% had disomy or nullisomy 13. These abnormal sperm were presumably due to that fraction of the gonadal tissue bearing the rob(13;13). Couple Both Heterozygous. An interesting curiosity is the extremely rare case of a union between Robertsonian heterozygotes. For example, MartinezCastro et al. (1984) describe two parents both with a 45,rob(13q14q) karyotype, whose three phenotypically normal children had a diploid number of 44, with their chromosomes 13 and 14 existing as a matching pair of rob(13q14q) translocations. Two rob(13q14q) × rob(13q14q) couples, being first cousin pairs and all four having the same rob(13q14q) by descent, each presented with three first-trimester abortions in Bahçe et al. (1996). A couple both carrying a rob(14q21q) are recorded having had a child with Down syndrome, with the unique karyotype 45,XY,rob(14q21q)pat,rob(14q21q)mat,+21mat (Rajangam et al., 1997). Similarly, Mori et al. (1985) reported a couple both of whom were 45,rob (13q15q), and who had had a child with translocation trisomy 13. Due to a founder effect, this
otherwise rare Robertsonian translocation was rather common in their small village in the province of Cuidad Real, in Spain, and this couple were surely distantly related, even though they were unaware of any link. The reader may care to construct a hypothetical balanced karyotype with 2n = 41 and five Robertsonian translocation chromosomes.
THE HOMOLOGOUS ROBERTSONIAN TRANSLOCATION (OR ACROCENTRIC-DERIVED ISOCHROMOSOME) This Robertsonian translocation chromosome comprises the long arm elements of two acrocentric chromosomes that are the same. The site of formation is typically postmeiotic (Robinson et al., 1994). If the translocation forms from the fusion of the two parental homologs, then manifestly there is biparental inheritance (Abrams et al., 2001). If, on the other hand, the rearrangement is actually an isochromosome, each long arm is an exact copy of the other, and there will be uniparental isodisomy. Such an isochromosome may have arisen as a “correction” of monosomy in the one-cell zygote. Rare cases of mosaicism for a “Robertsonian isochromosome” offer insights into causative mechanisms, albeit that these may not reflect the typical scenario. Bartsch et al. (1993) note some recorded cases of parental mosaicism for 47,+i(21q) and describe their own unique case of a woman with 47,+i(21p)/47,+i(21q)—some hundreds of cells from blood, gonad, marrow, skin were 47,+i(21p), and one single blood cell was 47,+i(21q)—who had had two children with Down syndrome due to the karyotype 46,i(21q). In herself, apparently, the isochromosomes arose as a postzygotic event from a 47,+21 conception, with classic centromere misdivision at the pre-embryo stage. The i(21p) line came to be the predominant in most tissues, but the i(21q) line had at least some representation in gonad and blood. DE TA I L S O F M EI OT I C B EH AV I O R
Only two segregant outcomes are possible at meiosis in the homologous 45,rob heterozygote. Either the gamete will receive the translocation chromosome, and be effectively disomic, or it will not, and be nullisomic. Essentially, this is 1:0 segregation (or “1+1”:0 segregation). No balanced gamete is possible. Thus, if the other gamete is normal, only trisomic or Robertsonian Translocations
•
149
monosomic conceptions are possible. Occasionally, conceptuses with translocation trisomy 13 are viable, and translocation trisomy 21 not infrequently survives to term. None of the other unbalanced possibilities (trisomies 14, 15, and 22,2 nor any of the monosomies) are viable. Postzygotic “trisomic correction” is a mechanism that, rarely, could enable the carrier to have a phenotypically normal child. If, say, in the case of an unbalanced 46,–22,rob(22q22q) conception, the free chromosome 22 were lost at a very early mitosis, genetic balance in this cell line would be restored, with a 45,rob(22q22q) karyotype. Provided the unbalanced cell line contributed negligibly or not at all to the embryo, and provided there were no effect due to uniparental disomy (and in the case of chromosome 22, there is not), the child would be normal. Very few such cases are recorded, with the 13q13q and 22q22q represented (Slater et al., 1994, 1995; Engel and Antonarakis, 2002; Ouldim et al., 2008). “Monosomic rescue” is another theoretical, and as yet unobserved mechanism in this context, whereby the homolog from the other parent could be duplicated postzygotically, as two separate homologs, or as an isochromosome, to produce a pregnancy with either a normal karyotype, or 45,iso. Finally, for completeness (but almost never in reality), gametic complementation is to be mentioned, whereby the non-rob parent contributes a gamete that happens to lack the homolog for which the rob parent’s gamete is disomic (Berend et al., 1999). For the rob(14q14q) and rob(15q15q) carrier, even
if one of these rescuing mechanisms did happen, the child would in any event be abnormal, since these UPDs lead of themselves to an abnormal phenotype.
GENETIC COUNSELING
THE HETEROLOGOUS ROBERTSONIAN TRANSLOCATION CARRIER I NF ERT I L I TY A N D M I S CA R R I A G E
The Robertsonian translocation involving nonhomologs is occasionally associated with repeated spontaneous abortion and with male infertility. The risks for miscarriage are set out in Table 7–4. It may be unclear, in an individual case, whether the association is causal or fortuitous. We can theorize that, in some miscarrying couples, there may have been a majority of zygotes with nonviable adjacent segregants; and in some infertile males, the translocation may have disrupted spermatogenesis. Cytogenetic analysis of products of conception, and of testicular tissue, respectively, may cast some light. It remains possible that some other cause could underlie the problem. The infertile male usually produces some sperm and may thus be a candidate for IVF using intracytoplasmic sperm injection (ICSI) and possibly PGD (Lee and Munné 2000) (and see Table 7–4 and Chapter 26).
Table 7–4. Risk of Miscarriage, and Proportions due to Trisomy, for the 13q14q and 14q21q Robertsonian Translocations, According to Gender of the Carrier rob(13q14q)
Mother Father
rob(14q21q)
MISCARRIAGE
TRISOMIC
MISCARRIAGE
TRISOMIC
22%–27% 13%
1%–7%* 1%
24% 33%
10%–14%* 1%
Note: It is apparent that translocation-related trisomy accounts for only a minority of the miscarriages. *Data depend on the gestational age at the time of ascertainment. Source: Table adapted from Kim and Shaffer (2002) and Engels et al. (2008).
2 In other settings, trisomy 22, extremely rarely, has gone through to stillbirth, or very short postnatal survival.
150
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Risks of Having Abnormal Offspring from Individual Translocations Figures for the risks to have an abnormal child, or for the probability of detecting an unbalanced form at prenatal diagnosis, are taken (making a few assumptions about extrapolating to the rare translocations) from data of a number of North American and European collaborative studies (Harris et al., 1979; Ferguson-Smith, 1983; Daniel et al., 1989) and set out in Table 7–2. These data relate essentially to the risk for a full trisomy. A potential risk of lowlevel residual trisomy, perhaps on the order of 1%, might suggest that a good number of amniocytes/ colonies be analyzed (Bruyere et al., 2001). Risks for UPD are drawn from the review of Shaffer (2006), again allowing for figures from the more common translocations being applicable to the rarer ones. Detailed comments on each individual translocation follow, with general comments thereafter on the theoretical risks of uniparental disomy, “isozygosity” for a recessive gene, residual low-level trisomy mosaicism, and interchromosomal effect. THE MO R E CO MMO N TRANSLO C ATIO N S
rob(13q14q). The karyotype of the balanced rob(13q14q) is shown in Figure 7–1. Translocation trisomy 13 can result from adjacent-1 segregation, with a typical Patau syndrome phenotype. The risk for this is very small. Almost all instances are index cases in families, not secondary cases. A review of several pedigrees in Harris et al. (1979), well subjected to statistical rigor, identified no increased risk for a malformed infant (they noted that a risk of up to 2% might have been missed, due to the sample size). In a European collaborative study, none of 230 prenatal diagnoses had an unbalanced karyotype (Boué and Gallano 1984), suggesting a risk of less than 0.4%. An incidence in Daniel et al.’s (1989) North American data of 3/204 (1.5%) may have been influenced by ascertainment bias, but in any event, combining the two data sets gives a figure of only 0.7%. A more recent study, based upon the impressive total of 101 pedigrees, provides support for these historic reports. Engels et al. (2008) identified no cases of translocation trisomy 13, after correction for bias, in live births, albeit that a number of their families had come to attention through an index case with translocation trisomy 13. These authors
propose risk estimates of <0.4% for female carriers to have a liveborn child with translocation trisomy 13, and <0.6% for the male, and if the genders are combined, one arrives at a lower figure of <0.23%. They did, however, document a 7% (3/42) incidence in amniocenteses. Further, one translocation trisomy infant had been stillborn; and it is a fine point, in undertaking this sort of analysis, to make a distinction between a stillborn baby versus one that survives only a few days (the usual in Patau syndrome). A risk estimate of ¼%–½%, or more conservatively <1%, may be a practical figure to offer. If there is male infertility, needing IVF with ICSI to achieve pregnancy, the additional exercise of PGD would be reasonable, to improve the chances of producing a normal/balanced conception; PGD may also be a reasonable choice for some female heterozygotes (see later). Otherwise, an offer of prenatal diagnosis remains a discretionary matter. A focused ultrasound should be capable of detecting the great majority of trisomy 13. The matter of UPD 14 has been noted earlier and discussed again later. rob(14q21q). The rob(14q21q) is the most important Robertsonian translocation in terms of its frequency and genetic risk, and it shows a marked difference according to the sex of the parent. Most familial translocation Down syndrome (DS) is due to the rob(14q21q) (Fig. 7–2). Adjacent segregation may lead to the conception of translocation trisomy 21 (Fig. 7–3). At amniocentesis, the female heterozygote has a risk for translocation trisomy 21 of about 15% (Fergusson-Smith, 1983; Boué and Gallano, 1984; Stene and Stengel-Rutkowski, 1988; Daniel et al., 1989). The risk of having a liveborn child with translocation DS is a little less (in the range 10%–15%): this likely reflects the loss, through spontaneous abortion, of a fraction of DS fetuses after the time during gestation when prenatal diagnosis is done. The risk for the male heterozygote is very different, and a figure of <1% is appropriate to offer. The matter of UPD 14 is noted later. T HE R A R E T R A N S LO CAT I O N S
rob(13q15q). Few data are available concerning genetic risks to the carrier (Mori et al., 1985; Daniel et al., 1989). We would expect these individuals are no more likely to produce adjacent segregants than the rob(13q14q) carrier, and a similar risk of <1% for translocation trisomy 13 may therefore apply. The risk for UPD 15 is noted later. Robertsonian Translocations
•
151
rob(13q21q). In Boué and Gallano’s (1984) study, the risk for translocation DS, in terms of the likelihood of detection at amniocentesis, was 10% for the female; and in Daniel et al.’s (1989) study, the figure was 17%. This 10%–17% range suggests there may be no real difference from the 10%–15% that applies to the common rob(14q21q). The risk for the male heterozygote is low, and probably similar to the <1% proposed for the male rob(14q21q) carrier. A 1% or less risk for translocation trisomy 13 may apply, for either sex. UPD is not a concern. rob(13q22q). We presume the risk for translocation trisomy 13 would be “small,” and perhaps similar to that for the rob(13q14q); a risk for trisomy 22 would presumably be minuscule.2 In Boué and Gallano’s (1984) study of 262 Robertsonian prenatal diagnoses not involving chromosome 21, there were only 3 rob(13q22q) cases, and in fact one of these showed trisomy 13; no unbalanced karyotypes were diagnosed in Daniel et al.’s (1989) seven cases. The man subjected to a sperm study in Anahory et al. (2005; see Table 7–3) had presented with infertility, and oligospermia was shown. UPD is not an issue. rob(14q15q). Adjacent segregants (translocation trisomy 14, translocation trisomy 15) are invariably lethal in utero. UPD 14 or UPD 15 are possible outcomes, as noted later. rob(14q22q) and rob (15q22q). The potentially trisomic states from these translocations (trisomy 14, 15, or 22) would all be anticipated to abort spontaneously.2 Neu et al. (1975) record the segregation of a rob(14q22q) chromosome in a large family, in which some carriers had an increased miscarriage rate. We comment later on UPD. rob(15q21q). From Boué and Gallano’s (1984) small series of nine carrier mothers, one (11%) had translocation trisomy 21 detected at amniocentesis; and in Daniel et al.’s (1989) data, the fraction was 0/9. These figures derive from too small a body of data to be sure, as yet, that the risk is really any different from the more solidly based 10%–15%, which applies to the rob(14q21q) female carrier. Again, we suppose a low risk (<1%) for the male carrier in terms of DS. The possibility of UPD is noted later. rob(21q22q). For a rob(21q22q) carrier parent, the risks for translocation trisomy 21 are about the 152
•
same as for the rob(14q21q), according to the sex of the parent. UPD need not be a concern, and neither, practically speaking, trisomy 22.2 U N I PA R EN TA L D I S O MY
UPD in a setting of parental Robertsonian heterozygosity is rare. We need consider only those acrocentric chromosomes subject to imprinting: chromosomes 14 and 15. The four syndromes that can theoretically arise are therefore UPD 14, maternal and paternal, and UPD 15, maternal and paternal. The UPD 14 syndromes are described in Engel and Antonarakis (2002) and in Sutton et al. (2003), and maternal and paternal UPD 15 are better known as Prader-Willi and Angelman syndrome, respectively. The potential mechanisms are, as discussed earlier, adjacent segregation followed by “correction” of trisomy with loss of a homolog, or (hypothetically) by “correction” of monosomy with replication of a homolog. In the review of Shaffer (2006), combining prenatal diagnostic data from seven groups, and including both familial and de novo cases, four instances of UPD were identified out of 482 prenatal diagnoses, for a point estimate, therefore, of 0.8%. The two familial cases were upd(13)mat due to a rob(13q14q)mat, and upd(14)mat due to a rob(14q22q)mat; and for the record, the de novo cases were upd(14)mat with a rob(13q14q), and upd(14)mat with a rob(14q21q). This pooled figure of 0.8% may possibly be a little less in the case of the father being the heterozygous parent (and possibly a little greater in the de novo case), but the numbers are too small to make that call, and it remains quite possible that no such differences exist. For practical purposes, the figure of 0.8% should be seen as applicable across the three parental classes (maternal, paternal, de novo), and across all types of rob. This risk is small, but not negligible. Thus, with respect to the “relevant” robs (those involving chromosome 14 or 15), it may be reasonable to consider adding UPD analysis to karyotyping, if the same 45,rob karyotype as the parent’s is observed. As for maternal and paternal UPD 13, 21, and 22, these are, apparently, without phenotypic effect, and need not be a cause for concern. “Isozygosity” for a Recessive Gene. Monosomic rescue, whether producing an isochromosome or a 46,N karyotype, theoretically has the potential to cause an autosomal recessive disorder, should the non-rob parent happen to be heterozygous for a
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
mendelian condition the locus for which was on the chromosome in question. But the risk is likely to be very low. In one small series in which a specific search was made for UPD due to monosomic rescue, from a rob parent, no such case came to light, and possibly it may never happen (Ruggeri et al., 2004). Barring knowledge of such a condition (e.g., Bloom syndrome, locus on chromosome 15) elsewhere in the family, molecular testing is not practicable. Of the more common recessive genes that might in some jurisdictions be suitable for population screening (cystic fibrosis, thalassemia, Tay-Sachs disease), none has its locus on an acrocentric chromosome. P RE I MP LAN TATIO N GE N E T I C DI A GNOSI S
In the series of Keymolen et al. (2009), a very impressive reproductive improvement was realized with the application of PGD. In 76 rob couples presenting for IVF, and with implantation having occurred (as measured by a positive hCG test), the take-home baby rate thereafter was 71%, compared with only 8% without the use of PGD. Furthermore, success was achieved within about a year, whereas the couples had been trying on average 3½ years before coming to PGD. They had mostly presented due to infertility (male carriers) and recurrent pregnancy loss (female carriers). Emiliani et al. (2003) point out the risk for associated mosaicism of the embryo and advise sampling of two blastomeres, rather than the usual single cell. Polar body biopsy (“preconception diagnosis”) is another approach available through a few IVF clinics, and obviously applicable only to the female heterozygote. Molina Gomes et al. (2009), in a pilot study, describe the procedure in seven women, six with 45,rob(13q14q) and one 45,rob(14q21q).3 From 32 embryos transferred, three successful pregnancies resulted. Interchromosomal Effect. The concept of an interchromosomal effect (ICE) has been invoked in the setting of the balanced Robertsonian heterozygote. Could a translocation somehow influence the distribution of another chromosome not involved in the rearrangement, with the production of a gamete aneuploid for a chromosome not involved in the translocation? Anecdotal reports of DS children
born to 14q22q and 13q14q rob carriers (Farag et al., 1987; Sikkema-Raddatz et al., 1997b) seemed to support this notion. However, formal segregation studies in large numbers of families with a rob(13q14q) or with trisomy 21 show no excess of trisomic offspring or of parental Robertsonian translocations, respectively (Harris et al., 1979; Lindenbaum et al., 1985). Therman et al. (1989) ascertained no Robertsonian translocation through a trisomic child other than one that included the trisomic chromosome. Sperm karyotypes of male heterozygotes show no excess of disomy for other chromosomes (Pellestor, 1990; Syme and Martin, 1992). These pieces of evidence amass a rather strong case that the Robertsonian translocation typically influences the segregation of no chromosomes other than those of which it is comprised. Against these observations, however, are those of Gianaroli et al. (2002), who examined the embryos produced at IVF from a number of rob heterozygotes, eleven male and four female, showing that imbalance due to common aneuploidies (mostly chromosomes 13, 16, 18, 21, 22) actually outnumbered those due to the translocation per se. Their privileged window of observation may have offered a clearer view, although, as indeed these authors acknowledge, the fact of infertility in the couples, and the need in many for ICSI to have been used, may have led to bias; other voices have spoken for this effect (Roux et al., 2005; Ogur et al., 2006; Chen et al., 2007c), and others unconvinced (Munné et al., 2005; Hatakeyama et al., 2006), while Baccetti et al. (2005) reiterate the point that ICE may be a consequence of the compromised spermatogenesis per se. In any event, the propositions that these other chromosomes as listed earlier should be included in the PGD test panel for the rob carrier, and that a case against an ICE is not proven, are to be noted.
THE HOMOLOGOUS ROBERTSONIAN TRANSLOCATION CARRIER We refer to these rearrangements as “rob” recognizing, as discussed earlier, that most such cases do actually involve an acrocentric-derived isochromosome (“rob-iso”). Virtually all conceptions of the heterozygote result in either trisomy or monosomy.
3 It was of interest that in five women where the infertility was due to the male partner, the imbalance rate was 30%, whereas in the two in whom a previous aneuploidy had been documented, the rate was 84%. Further studies will be necessary to confirm whether this finding might be more generally applicable.
Robertsonian Translocations
•
153
Monosomy results in occult abortion. Trisomy 14 and 15 always, and trisomy 22 virtually always, miscarry. Most trisomic 13 pregnancies miscarry, although some last until the third trimester; while of course many trisomic 21 pregnancies will proceed through to the birth of a child with Down syndrome. Practically speaking, no normal child could be produced from homologous Rob or isochromosome carrier individuals (the scenario of postzygotic correction, discussed earlier, can scarcely be raised as a realistic hope). Appropriate advice for these carriers is to consider sterilization. Alternatively, the use of donor gametes may allow the couple to have a normal child. Specific comments relating to the risk for abnormal offspring in each type of rob follow. rob(13q13q). The carrier parent can produce only monosomic or trisomic 13 conceptions, and these would either miscarry or, in the case of trisomy, produce a very abnormal child (Patau syndrome). Three recorded exceptions to this statement are given in Slater et al. (1994, 1995) and Stallard et al. (1995), of a normal parent having a normal child with rob(13q13q). The translocations were probably dicentric 13q isochromosomes, arising from postzygotic correction, and thus the children had uniparental isodisomy.4 rob(14q14q), rob(15q15q). Trisomies and monosomies for chromosomes 14 and 15 are not viable, and thus, all pregnancies of these heterozygotes would be expected to terminate in occult abortion or miscarriage. Even if postzygotic correction
did happen, the child would have a UPD syndrome, according to the translocation and the sex of the transmitting parent. Thus, it is, in theory and in reality, impossible to have a normal child from any gamete of the heterozygote. rob(21q21q). Although the rob(21q21q) is extremely rare, every counselor knows about this famous translocation. It is a classic example of a genetic risk of (practically) 100%. All pregnancies continuing to term can be expected to produce a child with DS. Sudha and Gopinath (1990), for example, report a couple who had 13 pregnancies, with 4 children proven or presumed to have had DS, and 9 miscarriages. The mother was 45,rob(21q21q). No case of postzygotic correction for this translocation has ever been reported. rob(22q22q). All conceptions would be monosomic or trisomic 22, other things being equal. For example, one carrier woman had 24 miscarriages, but no normal child (Farah et al., 1975). Two cases are mentioned earlier of postzygotic correction, with the birth of a normal child, but this is not a realistic hope to offer in the individual case. PREN ATA L D I A G N OS I S O F T H E D E N OVO HOMO LO G O US R O B ERTS O N I A N T RA N S LO CAT I O N
The de novo homologous Robertsonian translocation (or isochromosome) has a high risk for UPD; this entity is commented upon in the chapter on prenatal diagnosis (p. 470).
4 This makes the incidental point that UPD 13, maternal or paternal, is without phenotypic effect (Slater et al., 1995).
154
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
8 centromere fissions, complementary isochromosomes, telomeric fusions, balancing supernumerary chromosomes, and jumping translocations
THIS CHAPTER provides a setting for certain very rare abnormalities that cannot easily be accommodated elsewhere. Barely double-digit numbers, if that, of each category are known. Centromere fission results when a metacentric or submetacentric chromosome splits at the centromere, giving rise to two stable telocentric products. In a sense, this is the reverse of what happens in whole arm translocations. The heterozygote, a phenotypically normal individual, thus has 47 chromosomes. With the balanced complementary isochromosome carrier, two stable exactly metacentric products are generated. A balancing small supernumerary marker chromosome contains material deleted from the normal homolog. Telomeric fusion leads to a 45-chromosome count, due to the joining up of two chromosomes, tip to tip, not unlike the Robertsonian mechanism. The fusion chromosome has two centromeres, but one of these becomes inactivated. In jumping translocations, a segment can move from one chromosome to two or more recipient chromosomes. The Robertsonian fission reverses the fusion that had originally generated it.
BIOLOGY Centromere Fission In simple terms, a nonacrocentric chromosome undergoes a horizontal splitting at the centromere (Fig. 8–1a), although the true basis may be more complex than this (Rivera and Cantú, 1986; Perry et al., 2004). Two new telocentric chromosomes result (Fig. 8–2). One comprises the short arm of the original, and the other its long arm. It is as though the cell ignores the fact that the split happened and continues on normally, treating each part as a properly functioning whole. The other normal homolog remains intact. The heterozygous person (47,cen fis) may have a balanced complement of genetic material and thus be phenotypically normal. Among the few families on record, just six chromosomes—4, 7, 9, 10, 11, and 21—have been involved (Shim et al., 2007). The karyotype may be written, for example, 47,XX,–4,+fis(4)(p10),+fis(4) (q10). •
155
FIGURE 8–1 Comparing the processes of (a) centric fission and (b) complementary isochromosomes. The chromosome pairs at the outset (left) are to be imagined as existing in the zygote; they have replicated to give the double-chromatid state. The lightning arrow indicates misdivision of the centromere in one homolog. By the time the cell enters the first mitotic division (right), the abnormal states have been generated. Note that, according to the proposed mechanism in (b), uniparental isodisomy would necessarily result. Open indicates original homolog from one parent; crosshatched indicates original homolog from the other.
At meiosis in the heterozygote, the centric fission products presumably form a trivalent with the intact homolog, and 2:1 segregation, essentially as in the Robertsonian carrier, then follows. “Alternate” 2:1 segregation produces normal and balanced centric fission gametes, while adjacent 2:1 segregation leads to gametes disomic or nullisomic for either of the
7
7p
7q
7
7p
7q
FIGURE 8–2 Partial karyotype from a case of 47,cen fis(7). One chromosome 7 exists as a normal homolog, and the other homolog is represented by the 7p and the 7q chromosomes. 156
•
fission products (Fig. 8–3). Monosomy would probably be associated with occult abortion and trisomy with miscarriage or, in exceptional cases, with the live birth of an abnormal child. Thus far, trisomies for only 4p and 9p are on record. The paucity of data does not allow for a precise assessment of the genetic risk run by the centric fission carrier, other than to suggest it could, in some, be quite high. Dallapiccola et al. (1976) reported a chromosome 4 centric fission in a woman who had had two children with trisomy 4p and one normal child. Fryns et al (1980) describe a man and his normal daughter having a centric fission of chromosome 10. Recurrent miscarriage in the families of Janke (1982) and Shim et al. (2007) may well have been a result of asymmetric segregation of a chromosome 7, and a chromosome 11 centric fission, respectively; in the latter case, the cen fis 11 heterozygous woman then went on to have a normal 46,XX child. Miscarriages and childhood deaths in the family of Del Porto et al. (1984) might have been due to a cen fis 4, which was shown to have been
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
FIGURE 8–3 The six possible gametes arising from 2:1 segregation in a 47,cen fis(9) heterozygote. Two of these would be normal, the 46,N and the balanced 47,cen fis(9) states. Of the unbalanced states, only the 48,cen fis(9),+9p, in which the imbalance would be a 9p trisomy, might possibly be viable.
transmitted, in balanced state, from a mother to her son. Extremely Rare Complexity. A unique case in Fryns et al. (1985) offers a variation on the theme of centromere fission. A mother had deleted an interstitial segment of one of her no. 7 chromosomes, from the centromere to band q21 in the long arm, and this 7(cen-q21) segment existed as a free supernumerary chromosome. The remaining part of 7q, from q21 to qter, joined back on to the original centromere. Since the supernumerary chromosome was mitotically stable, it presumably included functional centromeric material. Her karyotype was balanced. She had had two severely malformed infants who inherited the deleted chromosome, 46,del(7) (cen-q21), but not the supernumerary 7q chromosome, in whom, therefore, the constitutional state was a monosomy for the interstitial segment.
be written, for example, as 46,XX,i(2)(p10),i(2) (q10). The usual mechanism of formation may be that, in the zygote, horizontal fission at the centromere of one homologous chromosome produces not two telocentric products (as happened in the fission, mentioned earlier), but two mirror-image metacentric chromosomes: an i(p) and an i(q) chromosome (Fig. 8–1b). This is followed by segregation of both isochromosomes into one daughter cell. There is loss (if it had ever been there) of the homologous normal chromosome contributed by the other parent (unlike the centric fission, in which the normal homolog is necessarily retained); thus, this is a form of monosomy rescue, which engenders a UPD, usually maternal (Bernasconi et al., 1996; Shaffer et al., 1997b; Björck et al., 1999). In other cases, one isochromosome may be of paternal origin,
Complementary Isochromosomes A not dissimilar case is the balanced isochromosome carrier: the “complementary short arm and long arm (p, q) isochromosome heterozygote.” Chromosomes 1, 2, 4, 7, and 9 have been reported with this picture, and at least four instances are known for chromosome 2 (Bernasconi et al., 1996; Shaffer et al., 1997; Albrecht et al., 2001; Baumer et al., 2007). The individual has a full complement of the chromosomal material—and may thus be phenotypically normal—but with the two p arms combined in one chromosome and the two q arms in the other (Fig. 8–4). A formal karyotype might
i(2p)
i(2q)
i(2p)
i(2q)
FIGURE 8–4 Chromosomes from a woman with complementary isochromosomes i(2p) and i(2q) (and see Fig. 8–1b). (Case of A. A Schinzel; Bernasconi et al., 1996.) Rare Abnormalities
•
157
and the other maternally derived, and this may reflect an initial trisomy rescue followed by postzygotic isochromosome formation (Kotzot et al., 2001; Albrecht et al., 2001; Baumer et al., 2007). A typical clinical presentation has been multiple miscarriage, in phenotypically normal women (Albrecht et al., 2001). Rather analogous to the rob(21q21q) carrier, it is practically impossible for such a person to have a normal child. Any pregnancies from “symmetric” segregation would be either dup(p)/del(q), or dup(q)/del(p), and thus hugely imbalanced.
Balancing Supernumerary Chromosomes If deleted material from a chromosome is then accommodated in a newly formed small supernumerary marker chromosome (sSMC), and if this extra chromosome can be stably transmitted, then the carrier individual can be of normal phenotype but may have a risk to have a child with a deletion, or a duplication, of the material in question (Baldwin et al., 2008, and see p. 209). The most remarkable example is that of a fourgeneration family, in which several persons carried a chromosome 22 with a classical q11 deletion, but this in company with a small supernumerary ring chromosome that comprised the deleted 22q11 material (Nevado et al., 2009). These people had, therefore, a balanced karyotype, and were phenotypically normal: 47,del(22)(q11),+sSMC. On classical karyotyping, the two chromosome 22 homologs had appeared normal, and it required fluorescence in situ hybridization (FISH) to reveal the deletion on one homolog; thus, the initial impression in this scenario may simply be 47,+sSMC, and the sSMC interpreted as “harmless.” In fact, two of these family members had had a child with typical deletion 22q11 syndrome. The other potential imbalance, that of dup(22q11) due to a 47,+sSMC karyotype, had not been observed in the family.
Telomeric Fusion This is the tip-to-tip fusion of two complete, or practically complete, chromosomes, and the person thus has a 45-chromosome count (Engelen et al., 2000; Lemyre et al., 2001). The fusion occurs at the level of the telomere or the subtelomeric region. All the necessary functional genetic material is “present and correct” (if there is a missing bit, it contains no 158
•
crucial genes), and the phenotype is normal, other things being equal. The composite chromosome has two centromeres (hence an alternative name of “stable non-Robertsonian dicentric chromosome”), but one of the two centromeres becomes functionally suppressed. The karyotype is written 45,t(A;B) or 45,dic(A;B), where A and B denote the two chromosomes. The short arm of an acrocentric chromosome is very frequently involved, and chromosome 18 is often one of the participating chromosomes. Ascertainment is typically fortuitous, or through reproductive difficulty (recurrent miscarriage, gonadal dysgenesis, oligoteratospermia). Familial transmission is recorded. The attachment of an essentially complete long arm of an acrocentric chromosome to the telomeric region of another autosome is a very similar circumstance (Fig. 8–5). A normal child could be produced following symmetric, essentially 2:1 segregation: that is, either the two normal homologs are transmitted or the composite chromosome. Asymmetric segregation, were it to happen, would lead to trisomy or monosomy of one of the component chromosomes, and, according to the nature of the chromosome, in utero viability would be compromised. For example, Lemyre et al. (2001) document a 45,XX,dic(14;18) (p11.2;p11.3) mother who was diagnosed, at 32 weeks gestation, with intrauterine fetal death. The fetal pathology examination was consistent with trisomy 18, and the karyotype, 46,XY,+18,dic(14;18)
FIGURE 8–5 A telomeric fusion translocation, 45,XY,t(8;15)(p23.3;q11). The normal father with this karyotype has all the functionally necessary part of chromosome 15 attached to the telomere of a chromosome 8. His child with Angelman syndrome has the same karyotype, but haplotyping with DNA markers showed that both chromosome 15 elements derived from the father, with no chromosome 15 contributed from the mother. Probably, this reflected a “corrected” interchange trisomy. (Case of A. Smith; Smith et al., 1994.)
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(p11.2;p11.3), confirmed this diagnosis. If the trisomic state were to be “corrected” by loss of the normal homolog from the other parent, a uniparental disomy would result. The case shown in Figure 8–5 is an example of this.
Jumping Translocation (“Translocation Sauteuse”) This evocative expression describes a mitotic rearrangement whereby the same piece of one chromosome breaks off, on more than one occasion, and attaches to the tips of other chromosomes. The site of breakage in the donor chromosome is characterized by the presence of an interstitial (internal) telomere, and this region offers the possibility of fusion with the recipient chromosomes. Only 26 constitutional cases were listed in the review of Iwarsson et al. (2009). Levy et al. (2000) identified the phenomenon in two couples, themselves karyotypically normal, presenting with recurrent miscarriage and showing evolving “jumping” cell lines in the cultured products of conception In one of these, for example, the conceptus was initially 46,XX,der(15)t(1;15) (q10;q10). A second line arose, with the 1q part of the der(15) replaced by an additional chromosome 15 which then generated an i(15q), along with (presumably independently) trisomy 7. Five further lines then budded off, all with considerable degrees of imbalance, the pregnancy eventually terminating in first-trimester abortion. Lefort et al. (2001) describe in some detail their own case, an otherwise normal boy with a (possibly coincidental) structural cerebellar defect. He had four separate cell lines, on blood and skin biopsy samples, with the segment 2p12-pter attached to 1pter, 5qter, 6qter, and 12qter, respectively. In each, the rearrangement appeared to be balanced. These authors proposed that these translocations were truly one-way, that is to say, having no reciprocal exchange, and with healing of the 2p12 stump by the formation of new telomeric sequences.
Robertsonian Fission The Robertsonian translocation is capable of reversing its evolutionary development, and the fused component chromosomes can separate. Perry et al. (2005) studied two families coming to attention due to a known family history of a segregating rob(13;15). They observed fission products, in
samplings of somatic tissues (chorionic villus, amniocytes, and blood) in 11 individuals or pregnancies, although mostly at single-digit percentage levels. These “new” acrocentric chromosomes were actually telocentric chromosomes 13 and 15, having no visible short arm material. This phenomenon appeared to be without any clinical consequence.
GENETIC COUNSELING Centromere Fission The centric fission heterozygote has a significant risk of having a phenotypically abnormal child in those cases in which a whole arm aneuploidy is viable. The 4p and 9p trisomies are the only examples known so far. It is most unlikely that any combination other than the short arm trisomy could be viable. Five percent to 25% is an educated guess of the likely risk range. Prenatal testing is certainly advisable. Of the phenotypically normal offspring of the heterozygote, half would be expected to have the centric fission and half to have normal chromosomes. For the heterozygote in whom neither whole arm imbalance is viable—an obvious example would be a 47,cen fis(1)—no risk for a liveborn abnormal child exists.
Complementary Isochromosomes In contrast, the carrier of the complementary p/q isochromosome carrier, essentially with certainty (that is, barring an extraordinary rescue event), cannot have a normal child.
Balancing Small Supernumerary Marker Chromosome The genetic risk is high, and it may approach 50%, if the del or dup imbalance implied by the material contained in the small supernumerary marker chromosome (sSMC) is “genetically small.” Nevado et al. (2009) emphasize the need to seek a cryptic deletion in persons found to carry a sSMC; if the true state of a cryptic deletion is not recognized, genetic advice would be gravely misplaced.
Telomeric Fusion Infertility may be frequent. If conception is possible, there is likely to be a substantial risk for aneuploidy of one or other of the chromosomes involved in the translocation, but equally, a normal child could be Rare Abnormalities
•
159
conceived. Uniparental disomy will need to be considered at least in the case of a chromosome 15 being one of the chromosomes.
the individual is unknown. The genetic implications for the next generation remain uncertain.
Robertsonian Fission Jumping Translocation These cases are typically de novo, and the reason for the chromosome suddenly becoming susceptible in
160
•
This appears to be a phenomenon of academic interest, seen only in somatic tissues, and of no clinical consequence.
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
9 inversions
INVERSIONS ARE INTRACHROMOSOMAL STRUCTURAL rearrangements. The commonest is the simple (or single) inversion. If the inversion coexists with another rearrangement in the same chromosome, it is a complex inversion. The simple inversion comprises a two-break event involving just one chromosome. The intercalary segment rotates 180°, reinserts, and the breaks unite (Fig. 9–1). The rearranged chromosome consists of a central inverted segment, and flanking distal, or noninverted segments. If the inverted segment includes the centromere, the inversion is pericentric; if it does not, it is paracentric. Figure 9–2 depicts two different pericentric inversions of chromosome 3. Note that the pericentric inversion has one break in the short arm and one in the long arm, whereas in the paracentric inversion both breaks occur in the same arm. Thus, when reading cytogenetic nomenclature, one can readily tell which is which: for example, 46,XX,inv(3) (p25q21) is pericentric and 46,XY,inv(11)(q21q23) is paracentric (inv = inversion). The clinical relevance of inversion chromosomes is that they can set
the stage for the generation of recombinant (rec) gametes that may lead to abnormal pregnancy. The heterozygote is, other things being equal, a phenotypically normal person. The reorientation of a sequence of genetic material apparently does not influence its function, and breakage and reunion at most sites do not perturb the smooth running of the genome. Some inversions of the X may be an exception to this rule: a breakpoint involving the X long arm within the “critical region” can cause gonadal insufficiency. Some pericentric breakpoints occur at preferential sites, including 2p13, 2q21, 5q13, 5q31, 6q21, 10q22, and 12q13 (Kleczkowska et al., 1987); and certain paracentric breakpoints are likewise overrepresented (Madan, 1995). CRYPT I C I N V ER S I O N S
An inversion may not necessarily be detected on routine study, and knowing when to mount a directed search requires clinical acumen. Thus, Yokoyama et al. (1997) discovered an inv(17) •
161
Normal Pericentric homolog inversion
Normal homolog
Paracentric inversion
FIGURE 9–1 The structure of the pericentric (left) and paracentric (right) inversions. The inverted segment is crosshatched. Asterisks provide landmarks at each end of the inversion segment.
(p13.1q25.1) in a father whose child had lissencephaly, a particular type of severe brain malformation. At first sight, the inverted chromosome looked normal. They noted a family history of similarly affected children, suspected a diagnosis of MillerDieker syndrome (which is due to 17p13.3 deletion), and went on to demonstrate the cytogenetic
abnormality using fluorescence in situ hybridization (FISH) with a Miller-Dieker chromosome 17 cosmid probe. Chia et al. (2001) studied a girl with an apparent del(2)(q37) on high-resolution analysis. Using subtelomeric probes to clarify the nature of the deletion, they were surprised to see a 2p signal at each end of the chromosome. Thus, the “deletion chromosome” could be seen for what it really was: a recombinant inversion chromosome, the essential genetic consequence of which was a deficiency of distal 2q. Since the short arm breakpoint was right at the tip of the chromosome at 2p25.3, there may have been little or no duplication of functional 2p genetic material. Microarray methodology was needed to clarify in detail the nature of a chromosome 20 inversion, in which classical karyotyping had been interpreted as normal, but with multiplex ligation-dependent amplification (MLPA) and FISH then revealing a del/dup of distal 20p/20q, respectively, in three adult siblings (Stevens et al., 2009). The mother’s karyotype was 46,XX,inv(20)(p13q13.33), and the siblings each had the identical rec(20)dup(20q) inv(20)(p13q13.33)mat. The imbalances were molecularly very small, the duplication being 2.5 Mb, and the deletion, 1.1 Mb. Dysmorphology was subtle, but the cognitive/behavioral phenotypes were quite abnormal. BREA K PO I N TS W I T H I N G EN ES
FIGURE 9–2 Two pericentric inversions of chromosome 3. Both of the noninverted segments are small in one (a) and one is large in the other (b). (Cases of N. A. Monk and L. M. Columbano-Green.) 162
•
In the event that a breakpoint occurs within a gene, the inversion could be directly pathogenic. Rare de novo examples include an inv(16)(p13.3q13) disrupting the Rubinstein-Taybi syndrome locus, an inv(17)(q12q25) disrupting SOX9 and causing campomelic syndrome, and a de novo inv(20) (p12.2p13) with one breakpoint occurring between exons 5 and 6 of the JAG1 gene, causing Alagille syndrome (Maraia et al., 1991; Lacombe et al., 1992; Stankiewicz et al., 2001b). A de novo inv(2) (q35q27.3) provided, in fact, the entrée to the mapping of a Waardenburg syndrome locus to 2q35 (Ishikiryama et al., 1989). Familial examples include an inv(15) (q11.2q24.3) transmitted from a normal mother to her Angelman syndrome daughter, and which led to the cloning of the UBE3A gene, mutation within which causes the syndrome in some subjects (Greger et al., 1997). Potentially, a familial inv(5) (p15.1q11.2) cosegregating with benign neonatal
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
convulsions might reflect a gene disruption (Concolino et al., 2002). In a family with a number of members suffering from attention-deficit disorder, and the affected persons also carrying an inv(3) (p14q21), a locus at each breakpoint was disrupted, these being in an intron of the DOCK3 gene at the p arm breakpoint, and in an intron of a solute carrier gene (SLC9A9) at the q arm (de Silva et al., 2003). One or other of these genes is a fair candidate for having a role in the genesis of this neurobehavioral disorder. On the X chromosome, a familial inv(X) (p11.4q22) damaging the Norrie syndrome gene is described in Pettenati et al. (1993). Xu et al. (2003) report a family with congenital androgen insensitivity (see p. 335) segregating an inv(X)(q11.2q27). Presumably, the break at Xq11.2 compromised the integrity of the androgen receptor locus. An inversion chromosome with gene damage at both breakpoints was reported in Saito-Ohara et al. (2002). A mother with the karyotype 46,X,inv(X) (p21.2q22.2) had a severely retarded 46,Y,inv(X) (p21.2q22.2) son with Duchenne muscular dystrophy, these effects being due to disruption of the dystrophin gene at Xp21.1, and of the RLGP gene at Xq22.2 (and there was also duplication of the proteolipid protein gene at Xq22.2). D E L E T IO N O R D UP LI CAT I ON AT I NVE R S IO N B REAK POI N T
A “clean” break and rejoin may not necessarily happen, and the rearrangement may, rarely, comprise, or give rise to, an associated deletion or duplication. Langer-Giedion syndrome (LGS) is due to a deletion at 8q24.11-24.13 (p. 317), and Sasaki et al. (1997) studied a child with LGS who had a de novo inv(8)(q13.1q24.11). Molecular analysis revealed a 4 Mb deletion encompassing the LGS region; presumably this segment had been deleted as part of the process that generated the inversion. A familial inv(18)(q21.1q23), in which a gene for brain myelination and presumably some adjacent genes were deleted, led to some features of the 18q–syndrome in a mother and daughter (Keppler-Noreuil et al., (1998). A familial inv(15)(p11q13), when transmitted from mother to child, underwent loss of the region that contains the putative Angelman syndrome (AS) locus (Webb et al., 1992). The loss was not detectable cytogenetically—the child appeared to have the same inversion that his mother and grandfather carried—but was revealed on molecular
analysis. The child had AS. Kähkönen et al. (1990) likewise describe a child with Prader-Willi syndrome and a 15q11 deletion whose father and grandmother were 46,inv(15)(p11q12) carriers. Lacbawan et al. (1999) reported a child who had inherited a paternal pericentric inversion of chromosome 2 that also had a de novo deletion at the 2p breakpoint, der(2)del(2)(p11.2p13) inv(2) (p11.2q13). The father’s inversion 2 is considered to be a normal population variant. The authors reviewed similar cases that occurred on chromosomes 1, 7, 13, 15, and 17 and raise the possibility that the deletions reported were directly related to the presence of the inversion. A deletion may be close to, but not actually a part of, an inversion breakpoint; and this then raises a question of a coincidental association versus a broader effect of the rearrangement (Lybæk et al., 2008). INVERSIONS IN ACROCENTRIC CHR O M OS O M ES
Additional complexities may arise when the pericentric inversion involves an acrocentric chromosome, because the nucleolar organizing regions (NORs) become located on the long arm. The first clue that a pericentric inversion is present is the finding of a nonstaining gap in the long arm (this can also be seen if a NOR has been inserted into another chromosome arm). The nonstaining gap can be further characterized/clarified with silver stain, or FISH using a probe to the ribosomal genes. Leach et al. (2005) describe a de novo case, 46,XX,inv(14)(p12q11.2). “Normal Variant” Inversions. “Inversions” having a breakpoint within the heterochromatic regions of chromosomes 1, 9, 16, and Y are frequently seen, and they are to be thought of as variants, not abnormal chromosomes (see Chapter 16). The most common inversion in humans not involving centromeric heterochromatin is the inv(2)(p11.2q13); just two recorded cases in the world are known of a possibly related pathogenic recombination (see p. 174). Other presumed harmless inversion variants include the following: inv(3)(p11q11) and inv(3)(p11q12), inv(3)(p13q12), inv(5)(p13q13), and inv(10)(p11.2q21.2). The inv(10) has been rather extensively studied by a collaborative group of five laboratories in the United Kingdom, who had, between them, 33 families available for investigation (Collinson et al., 1997). They found no excess Inversions •
163
of infertility or spontaneous abortion among carriers; and incidentally, all carriers of the inv(10) may be descendant from the same ancient Northern European heterozygote (Gilling et al., 2006). A similarly large collaborative Canadian study came to a similar conclusion with respect to the inv(2). FREQUEN C Y O F IN V E RSI ON S
Excluding these variant forms, inversions are a fairly uncommonly recognized rearrangement. Estimates of frequency range from about 0.12% to 0.7% (pericentric) and about 0.1% to 0.5% (paracentric) of individuals (Van Dyke et al., 1983; Kleczkowska et al., 1987; Worsham et al., 1989; Pettenati et al., 1995). With respect to the paracentric inversion, Madan (1995), commenting that these are “the most common form of chromosomal polymorphism found in nature,” suspects that many small examples remain undetected.
THE PERICENTRIC INVERSION BIOLOGY The Autosomal Pericentric Inversion D E TAILS O F MEIOTI C BE HAV I OR
The inversion heterozygote may produce chromosomally unbalanced gametes, and in consequence suffer reproductive pathology. The chromosomal
imbalance is a result of the formation of a recombinant (rec) chromosome. This is “aneusomie de recombinaison”—aneusomy due to recombination. Recombination occurs if there is, within the inverted segment, a crossover between the inversion chromosome and the normal homolog. Synapsis and Recombination. Classically, crossing-over follows the reversed loop model (Figs. 9–3 and 9–4) (Anton et al., 2005). This configuration of the bivalent allows as complete as possible alignment and pairing of matching segments of the inversion chromosome and its normal homolog (homosynapsis). One (or an uneven number of) crossover(s) within the inversion loop, between a chromatid of the normal homolog and a chromatid of the inversion chromosome, leads to the production of two complementary recombinant chromosomes. One of these has a duplication of the distal segment of the short arm, and a deletion of the distal segment of the long arm (chromosome c-c´ in Fig. 9–4); and the other way around in the other rec chromosome (d-d´ in Fig. 9–4). Thus, the conceptuses that result would have both a partial trisomy for one distal segment and a partial monosomy for the other, or vice versa. Typically, only one of these— the least monosomic—is ever viable. Consider the recombinant 7 due to a paternal inversion illustrated in Figure 9–5. There is a duplication of the substantial segment 7p14.2→pter, and a deletion of only the tiny segment comprising the distalmost sub-band of 7q (7q36.3→qter). The countertype form, having a monosomy for 7p14.2→pter (and trisomy 7q36.3→qter) would, we suppose, cause a miscarriage.
FIGURE 9–3 Inversion loop in meiosis, direct observation. Left, inversion loop in a mouse study. Right, spermatocyte study of a man with inv(6)(p22q22.2). (From de Perdigo et al., 1989, Correlation between chromosomal breakpoint positions and synaptic behavior in human males heterozygous for a pericentric inversion, Human Genetics 83:274–276. Courtesy Y. Rumpler; reproduced with the permission of Springer-Verlag.) 164
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Recombinant offspring (c-c’): 46,XY,rec(7)dup(7p) inv(7)(p14.2q36.3)
FIGURE 9–4 Inversion loop in meiosis, theoretical recombinant outcomes (based on the inv(3) shown in Fig. 9–2a). Both sister chromatids are shown. The inversion (centromeric) segment is crosshatched, the long arm noninverted segment is stippled, and the short arm noninverted segment is open. The four possible gametic outcomes following one crossover within the inversion loop are depicted. Chromosomes a-a´ and b-b´ are the intact homolog and the inversion, respectively; chromosomes c-c´ and d-d´ are the dup p and dup q recombinant chromosomes. Compare with the actual observation in Figure 9–3, right.
The cytogenetic nomenclature to describe the recombinant karyotype is straightforward. In the above case, for example, we have Parent: 46,XY,inv(7)(p14.2q36.3)
It is not necessary to put ‘dup(7p)del(7q)’—the complementary deletion is taken as read. More fully, the nomenclature is 46,XY, rec(7)dup(7p)inv(7) (pter→p14.2::q36.3→p14.2::q36.3→qter)pat. This complex twisting of the chromosomes to form a loop may not necessarily take place. In an inversion with a short inverted segment (Fig. 9–7a), a partial pairing may occur. Both distal segments, or sometimes just one, align in homosynapsis. The inverted segment and the corresponding part of the normal homolog either “balloon out” (asynapsis of the inversion segment), or lie adjacent but unmatched (heterosynapsis) (Gabriel-Robez and Rumpler, 1994; Anton et al., 2005). Thus, no crossing-over can happen within the inverted segment, and recombinant products do not form. Conversely, some inversions with long inverted and very short distal segments may undergo synapsis of the inverted segment only, with the distal segments at each end remaining unpaired (Fig. 9–7b). Recombination can occur in this setting. The quality of the chromatin may of itself have an influence. If both breakpoints are in G-light bands, the lack of homology is detected at synapsis, and the chromosomes respond by formation of a loop, achieving a complete homosynapsis. If, however, one or both breakpoints are in a G-dark band, nonhomology may not be recognized, and heterosynapsis is not prevented (de Perdigo et al., 1989; Ashley, 1990). In this state, recombination is suppressed ( Jaarola et al., 1998). With specific reference to some X inversions, it may be that they have a lesser propensity to engage in recombination within the inverted segment (Shashi et al., 1996). Sperm Studies Sperm studies in a small number of inversion heterozygotes give an indication of the frequency with which recombination happens, at least in male gametogenesis (Anton et al., 2005; Morel et al., 2007). Table 9–2 sets out the findings from a number of such studies. Initially, this work was done using the sperm-hamster methodology; in the late 1990s and 2000s FISH came to be used, and this approach allowed very large numbers of sperm to be analyzed. Dual-color FISH methodology, with one color (say, green) for the p arm and another (say, orange) for the q arm of the inversion chromosome, can show whether a sperm is recombinant. Sperm with nonrecombinant chromosomes would show one orange spot and one green spot. A recombinant chromosome with two orange spots would reveal Inversions •
165
Table 9–1. Autosomal Pericentric Inversions from the Literature Review of Ishii et al. (1997) Associated with the Birth of a Recombinant Offspring, Listed in “Numerical” Order CHROMOSOME
INVERSIONS
2 3 4 5
p25.3q33.3 p25q25a p15.32q35 p13q35 p15.1q33.3
p14q35 p15.1q35.1a
p23.07q25.13 p15q36
p15.1q36
p23.3q24.1 p11.2q25.2a
p12q25
p25q35 p25q23 p13q28 p13q33 p15q32 p15.3q35 p23q27 p14.2q36.3 p22q22 p23q22a p11q26 p15q24 p11q25 p13q24.3 p11q21 p12q14 p13q22 p11q25 p11q11 p13.3q13.33 p11.2q13.3 p13q13.33 p11q21.09 p11q21 p13q12.2
6 7 8 10 11 12 13 16 17 18 19 20 21 22
p13q23.3 p11q22 p13q21a p13.1q22 p13.3q25.1 p11.2q12.2
p12q13 p13q31
p12q13.3
p13q13.1
p11.2q22.1 p11.2q13.31
p12q22 p13q12a
p11.2q21.3
Notes: Inversions from 52 families, published over the period 1981–1995, are listed (references in Ishii et al., 1997, plus the case illustrated in Fig. 9–5; and more recent cases in Lagier-Tourenne et al., 2004, Mehra et al., 2005, Grange et al., 2005, Schluth-Bolard et al., 2008, Tagaya et al., 2008, and Stevens et al., 2009). a Reported in more than one family.
the dup(q)/del(p) state, while vice versa the dup(p)/del(q) chromosome would have two green spots. The longer the inverted segment, the more likely is recombination to happen. We can separate the studied cases into those with a long inversion segment (over 50% of the length of the whole chromosome), and those in which it is short. In five examples from Table 9–2 with longer inversion segments, inv(3)(p25q21), inv(6)(p23q25), inv(7)(p13q36), 166
•
inv(8)(p12q24.1), and inv(8)(p23q22), the proportions of dup(p)/del(q) and dup(q)/del(p) recombinant chromosomes were substantial: 31%, 38%, 24%, 38%, and 13%, respectively. No recombinants at all were seen in inversions with a short (or a very short) inversion segment: three “normal variant” pericentromeric inversions of chromosomes 2, 3, and 9, and an inv(20)(p13q11.2). Morel et al. (2007) offer this rule: a high risk of recombination applies when the inversion segment is over 50% in length; the risk is small when the length is between 30% and 50%; and no recombination appears to take place when the inversion segment comprises less than 30% of the chromosome. And in any event, even if recombination occurred in a small inversion segment, the recombinant chromosome would have such a large duplication and deletion that the risk of an abnormal live birth would, very probably, be negligible. An exception to this rule is given in the longsegment inv(17)(p13.1q25.3) carrier reported in Mikhaail-Philips et al. (2005). Of the 2000 sperm scored, 73% showed balanced segregants, and only 1.4% showed the classical dup/del recombinants. Fifteen percent had deletion only of 17p, which is the basis of Miller-Dieker syndrome (and this the diagnosis in two pregnancies fathered by this man). And similarly, recombination was rare in another inversion with a large inverted segment, inv(1) (p31q12): only 23 recombinants seen in 5966 sperm, a fraction of 0.4% ( Jaarola et al., 1998). This reflected a near-complete suppression of recombination. Different inversions in the same chromosome can have quite different ratios. Caer et al. (2008) looked at sperm from three men, with three different chromosome 8 inversions: p12q21, p12q24.1, and p23q24, respectively. With the p12q21 inversion, almost all sperm, 97%, were nonrecombinant, whereas the other two (with larger inverted segments) had 60% nonrecombinant. Concerning the common inv(2)(p11q13), Ferfouri et al. (2009) studied seven men presenting either with infertility, or during the course of a family study. Of just over 7000 sperm, 99.7% were nonrecombinant; the rate of aneuploidy otherwise did not differ from a control group. This work is interesting in proving that recombination can occur, even with this very short inverted segment; equally, the very tiny fraction manifesting recombination is to be noted. The fractions of each recombinant type are essentially the same. In the inv(8)(p23q22) listed in Table 9–2, for example, about equal numbers of
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Table 9–2. Sperm Analysis of 22 Autosomal Pericentric Inversion Heterozygotes REC INV SEGMENT SIZE (%) NONRECOMBINANT*
inv(1)(p31q12)** inv(1)(p31q12)** inv(1)(p36q32) inv(1)(p36.3q43) inv(2)(p11q13) inv(2)(p11.2q13) inv(2)(p23q33) inv(3)(p11q11) inv(3)(p25q21) inv(4)(p16q21) inv(6)(p23q25) inv(7)(p13q36) inv(8)(p12q21) inv(8)(p12q24.1) inv(8)(p23q22)** inv(8)(p23q22)** inv(9)(p11q13) inv(10)(p13q22.3) inv(12)(p11q23) inv(17)(p13.1q25.3) inv(20)(p12.3q13.33) inv(20)(p13q11.2)
30 30 81 95 10 10 71 5 60 42 80 65 31 61 62 62 16 47 51 89 84 51
100 99.6 83 68 99.4 100 61 100 69 99.2 46 75 97 61 88 87 100 97 91 73 80 100
DUP(p)/DEL(q)
DUP(q)/DEL(p)
0 0.25 9 12 0 0 20 0 14 0.8 19 7 1 20 6 6 0 3 4 0.8 10 0
0 0.13 7 19 0 0 18 0 17 19 17 0.4 18 6 7 0 4 0.6 8 0
Notes: Frequencies of recombinant (rec) and nonrecombinant (non-rec) chromosomes are shown as percentages. The size of the inversion segment, as a fraction (%) of the whole chromosome, is noted. Note that, as a rule, the larger the inversion size (especially >50%), the greater the fraction of recombinant forms. The proportions of the two recombinant forms from each inversion chromosome, dup(p)/del(q) and dup(q)/del(p), are very similar. *Whether normal or the inversion. **These two pairs represent the same inversion initially studied by the sperm-hamster test, and subsequently by FISH. Note how close the findings are. Source: From the review of Morel et al. (2007).
sperm showed the del(p)/dup(q) state (which is viable), and the dup(p)/del(q) state (which is not), 7% and 6%, respectively. Segment Content and Viability While a long inversion segment can set the stage for recombination, what determines the viability of the recombinant conceptus is the functional content of the noninverted (distal) segments. We speak of a “genetically small” content if the combined effect of a duplication and deletion does not cause lethality during the earlier part of pregnancy but allows development to proceed well through the pregnancy and possibly to live birth. Thus, only those heterozygotes
who have inversions with genetically small distal segments will ever have a chromosomally unbalanced phenotypically abnormal liveborn child. The inversion shown in Figures 9–2a and 9–5 illustrates this case. Inversion heterozygotes in whom one or both distal segments are genetically large (e.g., Fig. 9–2b) cannot have an abnormal recombinant child, although they may well have an increased risk for miscarriage. Any recombinants produced by such a person would impart a degree of imbalance that would be lethal in utero. Genetic content corresponds fairly well to chromosome length. In inversion families in which recombinant children have been born, the distal Inversions •
167
7 inv(7)
7
inv(7)
rec(7) 7
rec(7)
7
FIGURE 9–5 Pericentric inversion 7 in father (left) of an abnormal child with a recombinant 7 (right). The recombinant chromosome has a duplication of just over half of 7p, and a minuscule deletion involving the distalmost subband of 7q. The child has a triple amount of the segment p14.2→pter. The karyotypes are 46,inv(7)(p14.2q36.3) and 46,rec(7)dup(7p) inv(7)(p14.2q36.3)pat. (Case of S. M. White.)
(noninverted) segments together comprise, on average, only 35% of the total chromosome length; whereas in families having no known recombinant offspring, the figure is 62% (Kaiser, 1988). Nevertheless, if the distal segments comprise “genetically small” material, a larger fraction would not necessarily exclude a reproductive risk. Consider the inv(13)(p11q14) and inv(13)(p12q13), in which the distal segments comprise as much as 75% of the chromosome length. Although the imbalance in the recombinant is large in terms of haploid autosomal length, the result in the dup(q) form is, in effect, a partial trisomy 13 (the partial monosomy for 13p being without phenotypic influence). This is, of course, well known to allow intrauterine and postnatal survival. Similarly, an inversion in chromosome 18 can have distal segments that may be long relative to a short inversion segment, but they are still small genetically, and the dup+del combination can be viable (Schmutz and Pinno, 1986; Ayukawa et al., 1994). With specific reference to chromosome 4, Stipoljev et al. (2002) reviewed 20 reported familial cases and showed that recombinant forms have never been seen in those with smaller inversions, but frequently in the larger ones. As noted earlier, it is typically the case that only one recombinant form is ever viable. This is rather impressively illustrated in Allderdice et al. (1975) in a kindred with the inv(3)(p25q21). Numerous cases of known or suspected dup(3q) children have been born, but none with the countertype del(3q).
There is not even an increase in the miscarriage rate, suggesting that the del(3q) is lethal very early in pregnancy and causes “occult abortion.” Viability with both recombinant forms from the same inversion, the dup/del and the reciprocal del/dup, is infrequently seen. Kaiser (1984) records this only in the case of inv(5)(p13q35), inv(13)(p11q22), and inv(18)(p11q21), and Hirsch and Baldinger (1993) add an inv(4)(p15.32q35), as do DeScipio et al. (2010a, b) with respect to an inv(20)(p13q13.3). These five instances have this quality in common: the noninverted segments are very short. It is instructive to consider the inv(4) (p15.32q35) in Hirsch and Baldinger (1993), in which recombinant offspring could be del(4p)/ dup(4q) or dup(4p)/del(4q) (Fig. 9–8). The four separate segmental imbalances are all well known individually to be viable. Distal 4p is, of course, the basis of the Wolf-Hirschhorn syndrome; and distal 4p trisomy has syndromic, if not eponymic, status. The distal 4q segment is small cytogenetically (0.25% HAL) and functionally, and duplication1 and deletion are quite well tolerated. So the respective imbalances in the combined states—the del(4p)+dup(4q), and the dup(4p)+del(4q)— remain sufficiently small to be viable, at least much of the time. The index case, with the former imbalance, is a severely retarded child with a WolfHirschhorn phenotype; and an aunt, having the latter combination, had rather minor dysmorphism and mental retardation. The inverted segment is very long: 87% of the total length of chromosome 4. Therefore, a crossover within the inverted segment is, we assume, very likely to take place. Thus, the genetic risks to heterozygotes for this inv(4) is high. Two other reported families, with slightly different breakpoints (4p16.2/4q35.1 and 4p15.1/4q35.1, respectively), also demonstrate a high risk for imbalanced offspring, with both recombinant products observed (Dufke et al., 2000; Maurin et al., 2009). An even higher risk might apply to the inv(13) (p11q22) described in Williamson et al. (1980), in a family with several documented, suspected, or possible recombinant abnormal offspring. Here, the contribution of 13p imbalance to the two recombinant states—the del(13p)+dup(13q) and the dup(13p)+del(13q)—has no phenotypic effect, and the effective “single-segment” imbalances of
1 Duplication for a considerably longer segment, 4q31.3→qter, comprising 1.15% of HAL, is viable, as the children in the frontispiece photograph illustrate.
168
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
dup(13)(q22→qter) and del(13)(q22→qter) are each well known to be viable. Applying the principles of “private segregation analysis” set out in Chapter 4, the risk for a recombinant form in this family comes to a high 50%. We emphasize again the point that, while the length of the inverted segment may influence the likelihood of recombination happening, it is actually the combined genetic content of the distal segments that is the direct determinant of viability of the recombinant form. During the period 1981–1995, over 50 papers were published which reported the birth (or prenatal diagnosis) of offspring having a recombinant chromosome that derived from a parental pericentric inversion. In their review of this body of literature, and adding a family of their own, Ishii et al. (1997) determined the involvement of specific chromosomal segments. Figure 9–6, which is taken from their paper, depicts the combinations of dup+del genotypes that have been associated with viability. A few of these, which are shown asterisked, were identified at prenatal diagnosis, and in those with no known postnatal case, viability through to term remains unproven. A glance at the figure is enough to see that the gaps—that is, the noninverted segments—are generally longer, and usually a lot longer, than the sum of the lengths of the two inverted segments. This serves to illustrate again the point that inversions with large noninverted segments are, as a rule, the ones with the greatest genetic risk. It is also to be observed that the thick bars (representing duplications) are mostly longer than the thin bars (deletions), a reflection of the preferential viability of the least monosomic combination. The individual autosomal inversions from this review are recorded in Table 9–1. Inversions with very small distal segments may stretch the limits of cytogenetic detection (as also noted in the introductory section). Biesecker et al. (1995) describe an inv(22) with the long arm breakpoint in subtelomeric 22q, with the terminal 23–30 centimorgans of 22q now attached to 22p, which required molecular analysis with microsatellite markers and then FISH with a distal 22q cosmid probe for its identification. Due to the relative lack of G-band landmarks in 22q, and the normal variation that occurs with 22p, the defect was not recognized on a 450-band cytogenetic study. The mother
carrying this inversion would have had, presumably, a risk approaching 50% to have a further abnormal recombinant child. Another inv(22) of interest is that described in Boyd et al. (2005), 46,XX,inv(22) (p13q13.1). This rearrangement, in contrast, was very easy to detect, in that the inverted segment involved about half of the long arm and was attached to the particularly long stalk region of the short arm. The stalk region was probably2 the site of the meiotic recombination which gave rise to her abnormal child with 46,XX,rec(22)dup(22q) inv(22)mat. Effect upon Fertility. Uncommonly, the inversion heterozygote can be infertile (Groupe de Cytogénéticiens Français, 1986b; De Braekeleer and Dao, 1991). Abnormal synapsis of the chromosome pair can affect cellular mechanics at meiosis in the male, more likely if the inversion involves a larger chromosome, in consequence arresting spermatogenesis (Gabriel-Robez and Rumpler, 1994). Meschede et al. (1994), for example, describe azoöspermic brothers, one with histologically documented arrest at the level of the primary spermatocyte, and each heterozygous for an inv(1)(p34q23) inherited from their mother. Parental Mosaicism. Mosaicism for a (balanced) inversion is rare indeed. Lazzaro et al. (2001) describe a mother with 46,XX[11]/46,XX,inv(21) (p12q21.1)[19] on blood karyotyping, who had a child with a partial form of Down syndrome. The child’s karyotype was nonmosaic 46,XX, rec(21)dup(21q)inv(21)(p12q21.1)mat. Given the mother’s karyotype was from a peripheral blood sample, and she having had a recombinant child, clearly enough this is a case of somatic-gonadal mosaicism. Pericentric Inversions Frequently Innocuous. Many pericentric inversions are not associated with any discernible reproductive problems. The families of Voiculescu et al. (1986) and Rivas et al. (1987) are not atypical—an inversion chromosome transmitted through several generations, with numerous carriers identified, and no difference between the offspring of carriers, and those of noncarriers, in the incidences of abortion and neonatal death.
2 The mother’s normal homolog had a short stalk, the inversion a notably long stalk, and the recombinant chromosome in her daughter had a stalk of intermediate length.
Inversions •
169
FIGURE 9–6 Viable recombinants from 55 recorded parental pericentric inversion chromosomes. The pairs of bars alongside each chromosome ideogram, one thick and one thin, show the inverted segments. The thick bars indicate which is the duplicated segment, and the thin bars the deleted segment, in the recombinant offspring. The detail of the actual breakpoints is set out in Table 9–1. The inversions are grouped according to those chromosomes in which a dup(q)+del(p) is consistently seen in the recombinant offspring (above left), those in which a dup(p)+del(q) is consistently seen (above right), and those in which either pattern may be observed (below). Most of these recombinants had been reported in only one or a few cases, with the notable exception of the inv(8)(p23q22), observed on 54 occasions. Asterisks indicate that a case had been diagnosed prenatally; the inv(4)(p13q28) and the inv(5)(p13q33) had been seen only at prenatal diagnosis, so viability to term is not proven in these cases. (From F. Ishii et al., 1997, Case report of rec(7)dup(7q)inv(7)(p22q22) and a review of the recombinants resulting from parental pericentric inversions on any chromosomes, American Journal of Medical Genetics 73:290–295. Courtesy F. Ishii; reproduced with the permission of Wiley-Liss.)
Interchromosomal Effect Is Unlikely. Some pericentric inversions have been discovered in the setting of a child with an aneuploidy such as trisomy 21, and “interchromosomal effect” has been invoked (Groupe de Cytogénéticiens Français, 1985b). More likely, these associations are fortuitous: sperm studies endorse this inference (Anton et al., 2002; Mikhaail-Philips et al., 2004). It is intriguing to note that one case of cystic fibrosis due to maternal uniparental isodisomy 7 occurred in the setting of a maternal pericentric inversion variant for chromosome 7. This link seems more likely coincidental than causal (Voss et al., 1989). In one instance, “intrachromosomal effect” in an inv(21) is considered to have caused trisomic and monosomic 21 conceptions (Gabriel-Robez and Rumpler, 1994).
the observations in fewer than 30 families. The X inversion forms in the same way as an autosomal inversion, but the implications are different. This is because (1) breakpoints in certain parts of the X (its critical region) may have an influence on the phenotype of the female; (2) X chromosomal imbalance in the 46,X,rec(X) female may be mitigated by selective inactivation of the abnormal X; and (3) the 46,Y,rec(X) conceptus will have a partial X nullisomy and functional X disomy. The inv(X) can be transmitted both by the males and females. Baumann et al. (1984) and Schorderet et al. (1991), for example, describe families with an inv(X) transmitted through four generations, with all carriers—female heterozygotes and male hemizygotes—being phenotypically normal. The female and male inv(X) carrier need to be looked at separately.
RARE CO MP LEXITIES
Collectors of remarkable cases will find fascinating the report of Allderdice et al. (1991). They studied a kindred (mentioned also earlier) with a segregating inv(3)(p25q21), which originated from a couple marrying in 1817, and which was quite widely spread over the maritime provinces of Canada and other parts of Eastern Canada and the Northeastern United States. In the course of the study, a normal man was found to have two recombinant 3 chromosomes: one with a dup(q)+del(p), and the other with a complementary dup(p)+del(q), such that his karyotype was balanced. Probably, both of his parents were inv(3)(p25q21) heterozygotes, and one produced one recombinant gamete, and the other the other. Essentially the same scenario is described in Kariminejad et al. (2011): a consanguineous couple each heterozygous for inv(18) (p11.31q21.33) produced a child with a complementary recombinant karyotype, of normal phenotype, and in whom analysis showed segmental upd(mat) for 18p and segmental upd(pat) for 18q.3 A high prevalence of an inversion due to presumed founder effect is proposed in the Guadalajara region of Mexico, from whence there have been reported a number of cases of rec(22)dup(22q) due to a parental inv(22) (p13q12.2) (Tonk et al., 2004).
The Pericentric Inversion X Pericentric inversions of the X are rare indeed, and in 1997 Madariaga and Rivera were able to review
T HE F EM A L E I N V (X) H ET ER OZYG OT E
Outwardly, the female heterozygote appears normal, and not infrequently may be of normal fertility. The concept of “position effect” is of practical importance in the context of X rearrangement. If the long arm breakpoint lies within the segment Xq13→q22 or Xq22→q26, gonadal dysfunction may occur (Therman et al., 1990). There may be primary amenorrhea; or, after a fertile period in early adulthood, a premature menopause. Meiosis would be expected to proceed according to one of the preceding scenarios (see Figs. 9–4 and 9–7), with recombination within the inverted segment a possibility. While there is little practical experience to go on, we presume that an ovum with a normal X or the intact (nonrecombinant) inv(X) would produce a normal child, whether male or female. In the case of the male, this would require there to have been no compromise of loci at the breakpoints, and evidence of normality in the male in another family member would be reassuring. A hemizygous son would typically be of normal fertility (Madariaga and Rivera, 1997). If, in the family, the balanced inversion is associated with normal gonadal function in the female, a heterozygous daughter would be expected to have, likewise, normal puberty, fertility, and menopause at the usual time. This family information may not be accessible (or may not exist). In the family of Soler et al. (1981), for example, a hemizygous father had three sons and three daughters—each
3 These authors raise the intriguing theoretical point that continuing inbreeding in a region with a high prevalence of such a rearrangement could lead to several homozygous individuals being the beginning of a “new” species.
Inversions •
171
(a)
(b)
FIGURE 9–7 Alternative models for meiotic pairing, in which only a partial synapsis is achieved. Synapsis of (a) both distal segments; (b) the inverted segment. One crossover is shown in each.
Heterozygous parent
daughter, of course, an obligate heterozygote. He, apparently, had no gonadal deficiency; but his two older daughters had menopause at 37 and 34 (the youngest was only 30). There was no family history recorded antecedent to him. An ovum carrying a recombinant X would have two very different results, depending on whether it is fertilized by an X- or a Y-bearing sperm, as follows. The 46,X,rec(X) Conceptus. In their review, Madariaga and Rivera (1997) record outcomes in recombinant cases in 10 families. The del(Xq)/ dup(Xp) combination is, in female offspring, characterized by normal or tall stature, and ovarian dysgenesis. The countertype, del(Xp)/dup(Xq), is associated with short stature and, in some, intact ovarian function. These phenotypes presumably reflect the loss of stature genes (such as SHOX) located on Xp, and ovarian genes located on Xq, respectively. Any effect of the concomitant duplication is, presumably, mitigated by selective inactivation of the recombinant X chromosome. There is no obvious effect upon intellect.
Recombinant offspring del 4p & dup 4q
p15.32
q35
dup 4p & del 4q 4
inv(4) (p15.32q35)
rec(4) dup(4q)
rec(4) dup(4p)
FIGURE 9–8 An inversion inv(4)(p15.32q35) with small noninverted segments, in which each of the two recombinant possibilities is viable. The del(4p)/dup(4q) karyotype (left recombinant offspring) produces a Wolf-Hirschhorn-like picture, and in the dup(4p)/del(4q) case (right recombinant offspring) the phenotype resembles the partial 4p trisomy syndrome. The normal chromosome 4 contributed by the other parent is shown grayed out. The 4q segment is so small (indicated by the dots) that it might not to make a major contribution, whether duplicated or deleted, to the phenotypes. (From the case of Hirsch and Baldinger, 1993.) 172
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Consider the case presented by Buckton et al. (1981) (Fig. 9–9). One of the breakpoints is at the tip of the short arm, and the other is in proximal Xq. The recombinant chromosome, with a deficiency of the tip of Xp and a duplication of distal Xq (Fig. 9–9, lower right), was, in this family, associated only with shortness of stature. The partial Xq trisomy made no discernible contribution to the phenotype. A 26-year-old mother with the rec(X) herself had a rec(X) daughter: unarguable evidence that oögenesis had not (at least by age 26) been compromised. The 46,Y,rec(X) Conceptus. There will be a nullisomy for the deficient X segment. If this segment constitutes any but the tiniest length of chromatin, the conceptus would not be viable. Nullisomy for a tiny telomeric segment may be viable, but with major dysmorphogenesis and severe neurodevelopmental compromise. Furthermore, the concomitant disomy X is functional, not being subject to inactivation, and therefore of itself produces a
major deleterious effect (Groupe de Cytogénéticiens Français, 1986b). T HE M A L E I N V (X) H EM I ZYG OT E
In the male carrier, the rearrangement apparently has no effect on phenotype or on reproduction. Meiosis proceeds unperturbed (rather obviously, there can be no recombination within the inverted segment). All his daughters will be heterozygotes. Many will have normal gonadal function, although a family history of premature ovarian failure might predict the same problem (see also earlier discussion). Sons receive his normal Y and their mother’s (normal) X chromosome.
The Pericentric Inversion Y A pericentric inversion of the Y,inv(Y)(p11q13), is not uncommon in the general population (Verma et al., 1982; Tóth et al., 1984). It has no phenotypic
FIGURE 9–9 X chromosome inversion. The mother (above) has the karyotype 46,X, inv(X)(p22q13). Below, The two possible unbalanced reproductive outcomes in daughters, following recombination within the inverted segment; the normal X on the left in each has been contributed by the father. Each type of daughter would have a variant form of Turner syndrome. Male recombinant conceptuses are not shown: the combination of X nullisomy and functional X disomy in the 46,Y,rec(X) conceptus would in this instance be lethal in utero. (From the case in Buckton et al., 1981.) Inversions •
173
effect and implies no risk for having an abnormal child. It may be regarded as a normal variant. Meiosis proceeds as it would in the 46,XY male.
GENETIC COUNSELING The Autosomal Pericentric Inversion Variant Forms The not uncommon inv(2) (p11.2q13), a very small pericentric inversion, is practically always innocuous (Hysert et al., 2006; Ferfouri et al., 2009). Two possible exceptions are on record to belie its reputation: two abnormal children, one with a 2p duplication and the other a 2p deletion, the proximal boundary at or adjacent to 2p11.2, and the fathers being inversion heterozygotes, described as inv(2)(p11.2q12.2) and inv(2) (p11.2q13), respectively (Magee et al., 1998a; Lacbawan et al., 1999). It may be that the configurations adopted by the chromosome 2 homologs led to an unequal crossing-over, and hence the duplication or deletion. With only two such observations of recombination in the decades of history of clinical cytogenetics, some circumspection is required, and Lacbawan et al.’s comment that “at this point, it seems premature to recommend prenatal diagnosis of all couples in this situation” is reasonable. No genetic risks are known to be associated with the other inversion variants noted in the “Biology” section: “inversions” of 1, 9, 16, and Y heterochromatin, inv(3)(p11-13q11-12), inv(5)(p13q13), inv(10)(p11.2q21.2). Concerning the inv(10) (p11.2q21.1), Collinson et al. (1997) offer the practical advice that “family investigation of carrier status is not warranted in view of the unnecessary concern this may cause family members.” We exclude these inversion variants from the later discussion. RI SKS O F H AVIN G A N ABNO R MA L C H ILD
Ascertainment via Recombinant Child Identification of a family through a recombinant individual proves the viability of at least one of the two recombinant chromosomes. Table 9–1 lists a large number of different inversions for which a carrier is known to have had a recombinant child. There have been various empiric estimates of the overall level of risk to the heterozygote in families ascertained through an abnormal child. From a number of studies, a consensus range for the usual risk to have a liveborn abnormal child due to recombination is 174
•
5%–15% (Sherman et al., 1986; Stene, 1986; Groupe de Cytogénéticiens Français 1986b; Daniel et al., 1989). As a general rule, the longer the inversion segment—and, consequently, the shorter the distal segments—the greater the risk to produce a viable recombinant gamete. Very long inversions, such as that in Roberts et al. (1989), an inv(10) that comprised 80% of the whole chromosome, or the inv(20) in Stevens et al. (2009) comprising 94%, would imply the highest risks: in these particular cases, two out of the inv(10) carrier father’s three children were recombinant, and all three of the inv(20) carrier mother’s children. For the majority of families, there is probably no risk difference depending on sex of heterozygote (Kaiser, 1984; Stene, 1986); but in some families, the female heterozygote may run a greater genetic risk (Sutherland et al., 1976; Pai et al., 1987). Indeed, for the inv(21)(p12q21.1), recombinant children (with dup(21q) and thus a partial form of Down syndrome) have been seen only where it is the mother who is the carrier parent (Lazzaro et al., 2001). Each individual inversion carries its own individual risk. This may be arrived at by analyzing the patient’s family, studying the literature, and assessing the degrees of imbalance potentially arising in the recombinant conceptuses. A specific figure has been derived for one relatively common inversion, the inv(8)(p23q22): the risk for liveborn recombinant offspring, all of whom would have the del(p)/ dup(q) form, is 6.2%, for both maternal and paternal transmission (Smith et al., 1987). This compares closely with the figure of 6.9% of sperm with the del(p)/dup(q) form, attesting to an essentially uncompromised viability of the unbalanced embryo. In contrast, the countertype dup(p)/del(q) recombinant, which is seen in 6.3% of sperm, is never seen in liveborn offspring, reflecting zero viability. With inversions of 18, the breakpoints at p11 and at q11, q12, or q21, a “group risk” of 8% applies (Ayukawa et al., 1994). In due course, figures may be determined for other inversions seen in more than one family, such as the inv(3)(p25q21), inv(4)(p14q35), inv(10)(p11q25), inv(13)(p13q21), and inv(21) (p12q21.1). The risks to produce abnormal offspring from pericentric inversions in an acrocentric chromosome are again dependent on the size of the inversion, but in this case only the long arm segment needs to be considered; and rather than a composite del/dup imbalance, a recombinant chromosome would simply convey, in functional essence, either a
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
dup(q), for a partial trisomy, or a del(q), for a partial monosomy. A loss or gain of the p arm material would be without phenotypic consequence. The risk associated with a large inversion, with the q arm breakpoint sited distally, may therefore be particularly high; whereas a small inversion would, as typically, convey the least, and, for chromosomes 14 and 15, a practically zero risk (Leach et al., 2005). No Family History of Recombinant Form. For families identified by means other than through the birth of an abnormal child (e.g., discovered fortuitously at prenatal diagnosis), the overall risk is—for what this figure is worth—around 1%. The individual risk, which is what really matters, depends on the actual inversion. Is the inversion chromosome on record (Table 9–1) as being associated with viable imbalance? Or does the inversion segment include and extend beyond the inversion segment of one of these recorded cases? In that circumstance, a significant risk surely does apply (see earlier discussion). Is the inversion segment much shorter in length than any of those listed in Table 9-1? Here, the risk may be as low as zero. The level of risk can be assessed from a study of the family, noting the reproductive histories of other heterozygotes, and from a consideration of the degrees of potential imbalance in a conceptus. As a rule, any chromosome with a short inversion segment (less than one-third of the chromosome’s length) is most unlikely ever to lead to a viable recombinant product (Kaiser, 1988; Morel et al., 2007). Nevertheless, one should determine the composition of the theoretically possible recombinant gametes and gauge whether the resulting partial trisomy and partial monosomy might be viable. This applies in particular to inversions of chromosomes 13, 18, and 21, partial trisomies and partial monosomies of these chromosomes being well recognized as viable. If, in any inversion chromosome, one breakpoint is very close to the telomere, one recombinant form will impose very little partial monosomy. The contribution of the duplication can then be assessed on its own, and reference to the viability of this segment in other cytogenetic contexts (translocation, de novo rearrangement) will likely provide a valid comparison. For example, had the father in Figure 9–5 been identified before he had had children, we could have deduced that the rec(7) dup 7p genotype might survive to term, knowing that the databases of Stene and Stengel-Rutkowski (1988) and Schinzel (2001) record a viable phenotype for trisomy 7p14→pter.
Prenatal (or possibly preimplantation) diagnosis should be offered to the following individuals: (1) Any heterozygote in whose family a recombinant child has been born (2) A heterozygote for any of the inversions listed in Table 9–1 (3) A heterozygote for an inversion involving a segment longer than, but including, a region listed in Table 9–1 (4) Any other heterozygote for whom the theoretical recombinant product(s) might be viable. Many inversions of chromosomes 13, 18, and 21 will fall into this category. (5) Molecular analysis to exclude deletion in the Prader-Willi/Angelman region of 15q11-13 may be appropriate in an inversion having a breakpoint within or adjacent to this segment. Of the phenotypically normal offspring, approximately half will have normal chromosomes and half will be inversion heterozygotes (Groupe de Cytogénéticiens Français, 1986b). A risk to the child for some other rearrangement than the classic recombination (see earlier section “Deletion or Duplication at Inversion Breakpoint”) we presume to be very small, likely well under ½%, and prenatal array comparative genome hybridization targeted to the breakpoint regions would not normally be warranted. A question of interchromosomal effect appears not to be an issue.
The Inversion X The female heterozygote could have a premature menopause, if the long arm breakpoint is in the critical region, and if there is a family history of early ovarian failure; and practical advice might be to have children sooner rather than later. But normal reproductive function is perfectly possible. Recombination may be less likely than for an autosomal inversion (Pinto Leite and Pinto, 2001), although a risk to produce an abnormal daughter with a recombinant X does, nevertheless, exist. The abnormality is, to some extent, predictable according to the deleted segment, Xp or Xq: short stature is typically seen in del(Xp), and ovarian failure in del(Xq). Hemizygous sons would be expected to be normal, and reassurance in this respect may be drawn from the observation, if it can be made, of normality in a male relative. For the most part, no risk exists for having an abnormal son, because Inversions •
175
recombinant male conceptuses, having partial X nullisomy and disomy, would be nonviable. Only when the breakpoints are very close to the telomere is male viability possible, and such a child would have major physical abnormalities and mental retardation, probably severe. Due to this male lethality, the sex ratio of the offspring would be 1 male:2 females. All daughters of the male heterozygote would be inv(X) heterozygotes. Other things being equal, they will be phenotypically normal. If the long arm breakpoint is in the “critical region,” and if heterozygous female relatives have had ovarian deficiency (e.g., primary amenorrhea, premature menopause), they may develop the same problem. All sons would have a 46,XY karyotype.
The Inversion Y This inversion is generally considered a normal population variant of no clinical significance. It is self-evident that all the sons of the inv(Y) carrier will be, themselves, inv(Y) carriers. They are all normal and, other things being equal, have normal gonadal function. All the daughters would be 46,XX.
THE PARACENTRIC INVERSION BIOLOGY D E TAILS O F MEIOTI C BE HAV I OR
According to classical theory, the phenotypically normal heterozygote for an autosomal paracentric inversion will only have children who are karyotypically normal, or with the same balanced inversion. They cannot have viable unbalanced progeny. If a recombinant gamete is formed following a crossover in the inverted segment, the chromosome would be either acentric (lacking a centromere) or dicentric (Fig. 9–10). An acentric chromosome is never viable, since it lacks a point of attachment to the spindle fibers. The dicentric is generally considered a lethal impediment, being attached to spindle fibers pulling in opposite directions, with the chromosome thus suspended between the daughter nuclei at telophase and excluded from either cell. If the dicentric were to rupture, however, the possibility theoretically exists for a product (this might be, effectively, a dup+del chromosome) to enter the zygote, and to be viable. Alternatively, if the 176
•
FIGURE 9–10 Theoretical recombinant products from classical crossover in paracentric inversion. One is acentric (ace), and the other dicentric (dic). The inversion segment is shown crosshatched, and the different directions of crosshatching indicate the parts proximal and distal to the crossover point.
dicentric were to be included in the nucleus of a gamete, McClintock’s classical breakage-fusionbridge cycle might impose an eventually insuperable obstacle to continuing cell division, as the chromosome is tugged in two directions by its two centromeres in succeeding mitoses after formation of the zygote. The possible scenarios are more fully dealt with in Madan (1995). What are the findings on direct observation of gametes? In brief, recombination is scarcely ever seen. Anton et al. (2005) review the small total of five sperm studies, with inversion segments ranging from 6% to 32%. The fractions of recombinant sperm ranged from zero to 0.81%. The study with the largest number of cells analyzed (8158) had a recombinant rate of 0.03%, this particular rearrangement described as inv(4)(p14p15.3). The inversion with the largest inverted segment, inv(14) (q24.1q32.1), was analyzed by karyotyping in 120 sperm, and none of them showed a recombinant (Martin, 1999). Brown et al. (1998) analyzed 282 sperm from a man with a paracentric inversion, 46,inv(9) (q32q34.3), whose wife had had a number of miscarriages (they also had two children). Recombination was suppressed in the inversion segment; but, notably, of the five recombination events within the segment that were observed, each involved at least two crossovers. Brown et al. suggested the following mechanism. Synapsis, which starts at the telomere, advances along the chromatids, and then
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
encounters a region of heterosynapsis and “stalls.” This stalling allows an increase in recombination in the chromatid regions that are already synapsed. Synapsis eventually advances past the inversion segment and continues toward the centromere. But within the inversion segment itself, an “active search” for homology goes on, which may require the chromatids to take on a particular configuration (such as a microloop), and this may set up a hotspot for recombination. Only rare double recombinants from this setting would be able to form morphologically normal chromosomes, with sperm that would then be able to continue along their process of maturation; sperm with a single recombination would be acentric or dicentric. Meiosis in oögenesis commences during fetal life, and its study therefore requires access to fetal tissue. Cheng et al. (1999) analyzed ovarian tissue from a 19-week termination of pregnancy, in which a de novo inv(7)(q11.23q21.2) had been shown at amniocentesis. By using a FISH probe for the Williams syndrome critical region (WSCR), which is at 7q11.23, they could determine whether the inverted segments were aligned alongside each other (homosynapsis) or not (heterosynapsis). Most cells showed the chromosome 7 homologs lined up side by side, but with the WSCR signals off from each other: thus, the inversion segment was unaligned. A classical inversion loop was seen in only 10% of cells. This example, concerning a small inversion segment, offers an explanation for the rarity with which recombinant forms are seen: the necessary prerequisite of homosynapsis may not often be attained. RECOMB IN ATIO N / R EU NI ON WI T H VIAB LE P RO D UCTS
Classical theory remains valid in essence, some exceptions notwithstanding. The abnormal process of “U-loop recombination” (Feldman et al., 1993; Mitchell et al., 1994) is a mutational event, not a predictable consequence of a “normal” meiotic process (albeit in a chromosome that is abnormal). Reunitant may be a better word than recombinant. The crossover within the inversion loop, instead of continuing on in the same direction along the chromatid, reverses upon itself as a “U-loop.” The mechanism is illustrated in Figure 9–11. According to this construction, the resulting reuniting chromosomes would have either a duplication of that part of the inversion loop proximal to the crossover, and a
Father
18
inv (18)
18
inv (18)
Child
18
rec (18)
18
rec (18)
and
rec(18)'
rec(18)
FIGURE 9–11 Above, parent with paracentric inversion and child with recombinant (“reunitant”) chromosome. Father has paracentric inversion of 18q, inv(18)(q12.1q23). The inverted segment is shown crosshatched (crosshatching changes slope at q21.3). Child has duplication of the segment q12.1→21.3 on the reuniting chromosome (shown crosshatched) and deletion q21.3→q23. (Courtesy N. L. Chia and L. R. Bousfield.) Below, Proposed mechanism of U-loop exchange depicted; asterisk indicates point of U-loop. The position of the point of exchange within the inversion loop (in this case, q21.3) determines the nature of the imbalance. There is duplication of chromatin proximal to the crossover point (q12.1→q21.3), and deletion of distal chromatin (q21.3→q23), as in the child’s rec(18); and vice versa in the complementary product, rec(18)´. (An alternative interpretation is that the father’s rearrangement is a within-arm insertion of 18q, rather than an inversion, in which case the karyotype of the child would have been derived from recombination in the inserted segment.)
Inversions •
177
deletion of that part distal to it; or vice versa. A crossover (or, rather, chromatid breakage with abnormal reunion) at one of the entry points to the loop would produce a duplication alone, or a deletion alone. Feldman et al. (1993) review the inversion duplication (inv dup) chromosome, and notably, of the six familial cases on record, five may have been due to presumed U-type reunion from a maternal paracentric inversion. Chia et al. (1992) describe a case that quite probably reflected the same mechanism, a man with 46,inv(18)(q12.1q23) who had a child with a duplication/deletion 18q syndrome due to a presumed rec(18)(pter→q21.3::q21.3→q12.1::q23→ qter) chromosome, as shown in Figure 9–11. Another very similar inv(18) case is noted in Hani et al. (1995). In their exhaustive review, Pettenati et al. (1995) collected about a dozen similar cases. These cases represented offspring in 3.8% of their series of 446 paracentric inversions; but since all of these offspring were probands, and some we actually doubt were truly paracentric “reunitants,” we presume the actual reproductive risk due to U-loop reunion or other abnormal process would be a much smaller figure (Sutherland et al., 1995). Madan and Nieuwint (2002) pursue this question and show that indeed most “paracentric inversions” found through a recombinant child were really insertions. Classical theory needs also to accommodate the phenomenon of centromere suppression, which, extremely rarely, can allow the basically dicentric recombinant to function stably as, in effect, a monocentric. The chromosome attaches to the spindle fiber at only one centromere, the other being nonfunctional or suppressed; thus, no fusion-bridge cycle is initiated. “Extremely rarely” could, at this writing, be defined as four recorded cases from an autosomal paracentric inversion. Mules and Stamberg (1984) describe an infant dying as a neonate with a rec(14) whose mother had an inv(14)(q24.2q32.3); Worsham et al. (1989) studied in considerable detail a child with a rec(9) from a maternal inv(9) (q22.1q34.3); Whiteford et al. (2000) report a dysmorphic infant with growth and neurodevelopmental retardation and having a major heart defect, with the karyotype 46,XY,rec(15)(pter-q26.3::q11.2pter)inv(15)(q11.2q26.3)mat; and Lefort et al. (2002) describe an abnormal child in whose dicentric rec(14) chromosome one centromere could be demonstrated to have been inactivated, the mother’s karyotype being 46,XX,inv(14)(q13q32.2). These four cases share the features of a large inversion 178
•
involving most or almost all of a long arm, and with the short arms (14p, 9p, 15p, and 13p, respectively) being genetically “small”: in other words, the dup p+q/del q combination might not impose a lethal imbalance. Only in this setting, and if the dicentric chromosome were stable, could recombination cause an imbalance that would be viable and allow the birth of an abnormal child. A mechanism reminiscent of paracentric inversion U-loop reunion may be the cause of some isochromosome Xq Turner syndrome (Wolff et al., 1996). Two zinc-finger genes (ZXDA and ZXDB) in proximal Xp, just above the centromere, have about 98% homology and transcribe in opposite directions. In X-to-X synapsis in some meioses, a small inversion loop in proximal Xp might enable ZXDA (the more centromeric locus) in one Xp to match up with ZXDB on the other Xp, and vice versa. Then, a breakage and U-loop reunion between the two ZXD loci would generate an isodicentric chromosome Xqter → cen → ZXDA :: ZXDA → cen → Xqter. Similar events at other loci may underlie other Xq isochromosomes, or supposed Xq isochromosomes (Giglio et al., 2000). A minuscule number of cases of other sorts of viable recombinant offspring are known (Worsham et al., 1989). A dicentric recombinant chromosome, pulled in two directions, may rupture and yield a deletion. This may be the mechanism in the case in Courtens et al. (1998), in which a mother with 46,XX,inv(18)(q21.1q22.3) had monozygotic twins with a deletion of the segment distal to the inversion (q22.3-qter), and duplication of a small part proximal to it (q12.1-q21.1). In Fig. 9–10, although the scale is not right for this example, it could be imagined that a break occurred in the dicentric recombinant chromosome just above the lower centromere. An alternative mechanism is that the abnormal synapsis may set up a milieu that encourages some other type of rearrangement to form, such as excision of an inversion loop, and unequal crossing-over at the base of an inversion loop, a format initially proposed by Hoo et al. (1982). Yang et al. (1997) propose such a scenario in a family in which the index child had the deletion 46,XY,del(17)(p11.2p11.2), while the father and two aunts carried the paracentric inversion 46,inv(17)(p11.2p13). The deletion removed the
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Smith-Magenis region. This was “the first unequivocal demonstration by molecular analysis that a parent who carries a paracentric inversion is capable of having a viable child with an unbalanced monocentric recombinant chromosome.” Paskulin et al. (2011) relate a similar story, a child with a deletion in 7q31.32q33 (nt 112,552,898–135,714,528) at the base of the presumed meiotic inversion loop, from a maternal 46,XX,inv(7)(q11.23q33). South et al. (2006) describe a 46,XX,inv(5)(p13.3p15.3) parent who had a child with del(5)(p14.3) cri-duchat syndrome, which was likely due to a dicentric recombinant chromosome having ruptured in the middle of the inversion segment. Phelan et al. (1993) report the unique case of a father with an inv(9)(p13p24) having a child with a rec(9) containing a tandem duplication, which they propose came from breakage and reunion between sister chromatids within the inversion loop. Another unique case is that described in McClarren et al. (2006), in which an inv parent had a child with a deleted ring 22, leading to diGeorge syndrome. An inversion with a breakpoint in the vicinity of 15q12 may lead to a rearrangement that would cause Prader-Willi syndrome or Angelman syndrome, as mentioned earlier in the pericentric case. Microarray analysis enables a more precise view of possible recombination/rearrangement from a paracentric inversion, and we have applied this methodology in the laboratory in Spokane to two cases. First, in a child with developmental delay and dysmorphic features, the microarray study showed a single copy gain at 7q31.1q31.31 and a single copy loss at 7q31.33q32.3. The intervening sequence was present in two normal copies. Metaphase and interphase FISH confirmed both the gain and loss. Parental FISH studies were performed and identified a paracentric inversion in the mother. In a second case, we identified a single copy gain at 18q21.32q23. Metaphase FISH confirmed the presence of an additional proximal signal on one chromosome 18. Parental metaphase FISH studies were performed and again revealed an apparently balanced inversion in the mother. Some inversion carriers have been ascertained through their having had many miscarriages (Madan, 1995). In most of these, surely, the discovery was fortuitous. One family with an inv(10) was widely studied, and 19 carriers in three generations had only one miscarriage out of 36 pregnancies (Venter et al., 1984). The report in Devine et al. (2000) mentioned earlier of two brothers with 46,XY,inv(2)
(q14.2q24.3) presenting with reproductive pathology may be suggestive, but other causes are quite possible. One brother’s wife had three miscarriages, and at in vitro fertilization in the partner of the other brother, five of ten fertilized eggs failed to cleave, and progression in the remaining five failed at the blastocyst stage. No karyotyping was done of any of these several products of conception. In a very few cases, theoretical dicentric recombinant products might convey a genetic imbalance that could allow at least some weeks of in utero growth before miscarrying (Bocian et al., 1990; Bell et al., 1991). The nine miscarriages suffered by the carrier grandmother in the family in Worsham et al. (1989) we might more reasonably imagine to have been due (some of them at least) to recombinant gametes, the dicentric state having been proven in her index grandchild. Coincidental Abnormality. In some instances, the finding of a chromosome abnormality in a child from a paracentric inversion parent may be coincidence, even if the same chromosome is implicated, as seen in the case reported by Bourthoumieu et al. (2003) of a child with cri-du-chat syndrome and a del(5)(p14pter) karyotype. Parental chromosome studies revealed that the mother carried an apparently balanced paracentric inversion of long arm of one chromosome 5. The authors concluded that the two rearrangements were unrelated. Other Mechanisms Causing Abnormality. Mendelian loci can be vulnerable when chromosomal rearrangement happens, due to “position effect,” epigenetic influence, or direct disruption. We have seen, for example, a family in which a chromosome 7 inversion, inv(7)(p22.2p21.2), has been associated with Saethre-Chotzen syndrome, this being a Mendelian disorder due to the TWIST gene. Two heterozygous children showed major craniosynostosis, but the father and grandfather had only the subtlest facial, auricular, and digital signs. It may be that Saethre-Chotzen syndrome due to position effect has a milder phenotype than in the case of point mutation (Rose et al., 1997). On chromosome 7, at 7q21q22, an inversion has been described in association with the split hand/foot malformation, and again it is proposed that “position effect,” with compromised expression of a putative hand/ foot morphogenesis gene(s) in neighboring chromatin, might be the underlying causal mechanism (van Silfhout et al., 2009). Another possible epigenetic Inversions •
179
example: Norman et al. (1992) described a family in which a mother had one child with Beckwith syndrome and a presumably affected fetus, all three carrying an apparently balanced inv(11) (p11.2p15.5). The normal imprinting state of the Beckwith region on distal 11p may have been perturbed. A Special Case, inv8p23. A paracentric inversion of 8p23 is a very common “abnormality,” and indeed should be described as a polymorphism, since it occurs in around a quarter to a third of European and Japanese populations, respectively. The inversion cannot be seen on classic cytogenetics and requires FISH with probes recognizing sequences within band 8p23 for its delineation. From the millions, perhaps billions of people who carry this inversion, the tiniest number of abnormal infants have been born, perhaps 50 or so known worldwide. In these very rare cases, the inversion has led to a classic recombination with production of a dicentric chromosome, essentially as outlined in Figure 9–10, in which a segment including one centromere is then “clipped off,” to produce a monocentric inv dup del(8p)(8qter→8p23::8p23→proximal 8p). The recombinant chromosome is typically generated in (unusually for a structural rearrangement) maternal meiosis (Shimokawa et al., 2004). X Chromosome. If a paracentric inv(X) is associated elsewhere in the family with normality, no defect would be anticipated in future heterozygotes or hemizygotes (Neu et al., 1988a). Breakpoints in the critical regions in Xq might, however, compromise gonadal integrity. For example, Dar et al. (1988) report a woman with a de novo inv(X) (q13q24) who had ovarian dysgenesis with primary amenorrhea and no spontaneous pubertal development, and Németh et al. (2002) describe an infertile man with a Klinefelter-like phenotype having an X inversion with rather similar breakpoints, 46,Y,inv(X)(q12q25). A woman with a somatotype of Turner syndrome having an Xp inversion (p11.2p22.1) is described in Dahoun (1990). A breakpoint might damage a mendelian locus, and Briault et al. (1999) report a family in which the FG syndrome (mental defect, facial dysmorphism, hypotonia, anal abnormality) cosegregated with a paracentric X inversion, inv(X)(q12q28). One FG locus has been mapped to Xq12-q21.31, and so it is plausible that the q12 breakpoint in the inversion may have been at the site of the FG gene. 180
•
Y Chromosome. Only two paracentric inv(Yq) cases are on record (Madan, 1995; Liou et al., 1997). In Liou et al.’s three-generation family, the normal grandfather and father were 46,X,inv(Y)(q11q21), and the child with the same karyotype had ambiguous external genitalia with Müllerian structures internally and intra-abdominal testes. The inversion Y may have been coincidental; alternatively, there may have been, in the child, a position effect whereby the expression of a gonadogenesis gene had been compromised. PARACENTRIC INVERSIONS U SUA L LY I N N O CUO US
The above rather extensive compendium notwithstanding, the observed facts attest to the general innocuousness of the autosomal paracentric inversion, concerning either the heterozygous state per se, or a risk for chromosomally unbalanced offspring. Madan (1995) reviewed 184 cases of autosomal paracentric heterozygosity. Many were ascertained fortuitously, and including those discovered during the course of investigation for recurrent miscarriage, 58% were identified in a normal person. Several had an abnormal phenotype, but this was, of course, the reason they had the chromosome test done in the first place: by definition, they had to be abnormal. No clear consistent pattern among phenotypes of presenting cases is apparent. As Madan comments, there may have been a bias in choosing cases for publication, and editors of journals might not find compelling a paper describing an “uninteresting” inversion discovered incidentally in a normal individual (a series of a dozen or so cases might stand a better chance). In their review, Pettenati et al. (1995) could be confident about a causal association with a specific phenotype only in the paracentric inv(X), and not with any of the autosomal inversions. The Groupe de Cytogénéticiens Français (1986a) note that the reproductive fitness of heterozygotes in 32 French families was normal. Two quite common inversions seen in a number of families in more than one part of the world are the inv(3) (p13p25) and the inv(11)(q21q23) (Madan, 1995). No abnormalities directly attributable to these inversions have been documented. It may be founder effect, or recurring mutation, that is the basis for their frequency. In the one sperm study of a paracentric inversion heterozygote, having the relatively common inv(7)(q11q22), Martin (1986) found no recombinants. The smallness of the inversion
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
segment may have been a factor militating against formation of a synaptic loop. (It is not without interest to note that a similar inversion is the norm in the gorilla chromosome 7, and so this human form could be thought of as a “back mutation” to that of the ancestral primate.) Interchromosomal Effect Is Unlikely. Watt et al. (1986) raise the possibility that the paracentric inversion might have an “interchromosomal effect.” They note an apparently high level of reported associations, within families, of an inversion plus some other chromosomal defect. We suspect this is artifactual; as these authors note, ascertainment and publication biases are potential confounders in this setting. Pettenati et al. (1995) reached a similar conclusion. Technical Comment. Paracentric inversions can be technically difficult to detect on classical cytogenetics. Gross chromosome morphology is not altered, and unless major landmark bands are shifted, the rearrangement may go unnoticed. Only with the use of good quality, high-resolution banding are paracentric inversions likely to be detected regularly. These cytogenetic difficulties may be why relatively few cases of this type of inversion have been published. Also, for technical reasons, reported cases of recombination in the literature should be regarded with caution; as mentioned earlier and noted later, some “inversions” are likely actually to be intrachromosomal insertions (“paracentric shifts”). The cytogenetic distinction can be difficult to make, especially so for chromosomal regions without distinctive banding patterns, or where the inverted segments are very small (Callen et al., 1985; Madan, 1995). For example, the inverted insertion of chromosome 15 described in Collinson et al. (2004), associated with recombinant offspring having Prader-Willi and Angelman syndrome (and see p. 364), had originally been reported, some 10 years prior, as a paracentric inversion. We have seen a family in which the index case seemed to have an unbalanced translocation at distal 4p, but the normal mother and grandfather had the same anomaly, which could then be reinterpreted as the minimum inversion detectable on routine cytogenetics, a one-band paracentric inversion, in this case inv(4)(p15.3p16.3) (Smith et al., 1992).
GENETIC COUNSELING On practical grounds, the reassuring point to note is that practically all paracentric inversion heterozygotes
identified have been discovered fortuitously, and not through the birth of a child with an abnormality attributable to the parental inversion (Madan, 1995). We agree with Madan: “the vast majority of paracentric inversions are likely to be harmless.” Apparently, the genetic risks to offspring are extremely small. In the U.S. collaborative study described in Daniel et al. (1988), there were no unbalanced karyotypes in 30 prenatal diagnoses. The sex chromosomes warrant separate attention, and it may be that some X and Y paracentric inversions have an effect upon gonadal development in the intact (that is, unrecombined) state. However, a tiny handful of abnormal offspring, and as reviewed at length earlier, refute a complete harmlessness in the parental paracentric inversion, whether due to classic recombination or to other forms of rearrangement. Whether this would warrant prenatal diagnosis, when a parent is a carrier of one of these inversions, or indeed for any paracentric inversion, is a matter for debate. Even where the new chromosome from a classic recombinant or U-loop reunion might on theoretical grounds be viable, the risk for one to be generated, while its exact magnitude is unknown, is surely “extremely small.” “Better than 99.9%” might be a fair estimate that there will be no untoward reproductive outcome due to behavior of the inversion. Albeit that microarray may offer the potential to screen, at prenatal diagnosis, for submicroscopic molecular damage associated with a particular apparently balanced inversion, the case for so doing is very modest. Nevertheless, it would scarcely be realistic, at the present time (although the future may hold a different scenario), to screen, at prenatal diagnosis, for submicroscopic molecular damage associated with a particular apparently balanced inversion. Caution should be exercised during genetic counseling, in that it is prudent never to say “never,” and no risks are ever zero. Thus, we suggest that, in practice, an offer of prenatal diagnosis be discretionary, in the case of a fortuitously discovered inversion in the family; and we would regard it as not inappropriate if the offer were declined. A firmer stance may be appropriate if there has been a previous history of an apparently associated reproductive abnormality. Inversions on record with a demonstrated recombinant would oblige the offer of prenatal diagnosis. These include those noted earlier: inv (7)(q31.31q31.33), inv(9)(p13p24), inv(9)(q22.1q34.3), inv(14)(q24.2q32.3), inv(17) (p11.2p13), inv(18)(q12.1q23), inv(18) Inversions •
181
(q21.1q22.3), and inv(18)(q21.32q23). But again, we return to the expressions used above: “vast majority”, and “better than 99.9%”, in respect of favorable behavior of the inversion. As mentioned earlier, a diagnosis of a paracentric inversion might be incorrect, and the rearrangement is actually a within-arm insertion, which carries a high genetic risk (p. 194). Since the distinction in the routine laboratory can be difficult, a practical view might be to risk overinterpreting subtle paracentric inversions as potential insertions, in those cases where the cytogeneticist is not absolutely certain. The true picture may emerge by determining the order of a number of FISH probes across the relevant region.
earlier, arises from a maternal cryptic (on classical cytogenetics) paracentric inversion. Yet for a couple who have had this happen, the risk of recurrence is still, in all likelihood, extremely small. Nevertheless, it would be understandable for a couple having had that experience to seek the reassurance of prenatal diagnosis in a subsequent pregnancy. The Paracentric Inversion Detected Prenatally. If an apparently balanced paracentric inversion is discovered at prenatal diagnosis, and if the parental karyotypes are normal, there yet remains a possibility that the rearrangement is not truly balanced, and a risk for abnormality exists. This question is dealt with in detail in Chapter 27 (p. 471).
The Special Case of the inv dup del(8p) The inv dup del(8p), noted in the “Biology” section
182
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
10 insertions
INSERTIONS ARE A TYPE OF TRANSLOCATION: sometimes the expression “insertional translocation” is used. In the common, simple insertion, three breaks are required. The first two breaks release an interstitial segment of chromosome, which is then inserted into the gap created by the third break. In the simple one-way interchromosomal insertion, a segment from one chromosome is intercalated into another chromosome. A more complicated four-break rearrangement is the reciprocal insertion, whereby two nonhomologous chromosomes exchange intercalary segments. In the intrachromosomal insertion, a segment is intercalated into another part of the same chromosome. The segment may be inserted “right way around”—that is, with the same orientation to the centromere as before; this is a direct insertion (dir ins). Or it may be reversed—an inverted insertion (inv ins). More complicated scenarios, which may involve both insertional and terminal translocated segments, are
more appropriately dealt with in Chapter 12 (Complex Rearrangements). Insertions are rare rearrangements, at the level of detection according to classical cytogenetics. With microarray technology, previously undetectable insertions of very small size are coming to light, de novo and familial, and it may prove that “uncommon” will be a more accurate adjective to describe frequency than is “rare.” Kang et al. (2010) found a 20-fold increased discovery of insertions, compared with earlier studies, in a large series of cases presenting with typical chromosomal clinical pictures. Many of these turned out to be (probably) harmless polymorphisms (CNVs), upon the recognition of a parent carrying the same insertion. But a fraction were, in all probability, truly pathogenic. It is notable that most of these would not have been detectable on classical cytogenetics. In this chapter, we consider the case of the phenotypically normal heterozygote, in whom the rearrangement is assumed to be balanced.
•
183
THE INTERCHROMOSOMAL INSERTION BIOLOGY The simple one-way interchromosomal insertion is the most common form of this uncommon rearrangement: Van Hemel and Eussen (2000) estimate a prevalence, on classical cytogenetics, in the order of 1 in 80,000. The formation of the rearrangement is depicted in Figure 10–1. The recipient chromosome now carries the insertional segment, and the donor chromosome lacks it.
thrown into a loop1 to accommodate this requirement (Fig. 10–2, upper). (Some crossing-over will presumably occur between synapsed regions, but this would not alter segregation outcomes.) Alternatively, homologs may pair along their full lengths, which would bring some nonmatching segments “incorrectly” alongside each other (“heterosynapsis”). Then, with normal segregation of the two bivalents, independently of each other, two alternative pairs of gametes are possible. Overall, there would be gametes of four possible segregant types, in the ratio 1:1:1:1: two with a correct amount of genetic material, and two without. The former two combinations are 46,N and the balanced insertion carrier.
D E TAILS O F MEIOTI C BE HAV I OR
In theory, two categories of meiotic behavior are possible, according to whether the homologs pair independently, or as a quadrivalent. I NDE P EN D EN T SY N APSI NG OF HOMO LO G O US PAIR S
Synapsis at meiosis I
Meiosis could proceed in the usual fashion, with homologs pairing independently as bivalents. In essence, we can suppose that the insertional segment is disregarded and that the homologs synapse, with segments matching for as much of their length as they are able. In theory, and perhaps only with larger insertions, the insertional segment could be
Normal Donor Recipient
Direct insertion
Inverted insertion
FIGURE 10–1 The formation of an interchromosomal insertion. Single and double asterisks indicate orientation of the insertion segment. The direct insertion has the same orientation to the centromere; the inverted insertion has the opposite orientation.
Balanced
•
Deletion
FIGURE 10–2 Gamete production following independent pairing of the two sets of homologs. The insertional segment is shown in black, both in its original and in its translocated positions. The horizontal line marks the site whence came the segment from the donor (crosshatched) chromosome, and the site of its destination on the recipient (white) chromosome.
1 Described also as ballooning out, looping out, folding out, or translocation loops.
184
Duplication
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
The two unbalanced combinations would produce conceptuses one with a partial trisomy (duplication) and the other with a partial monosomy (deletion), for the insertional segment (Fig. 10–2, lower). As discussed later, studies of testicular biopsies and sperm have shown that (at least with smaller insertions) the homologs pair normally as bivalents, and that the expected ratios hold true. It makes no difference whether the insertion is direct or inverted. The foregoing scenario of independent synapsing is more likely to apply when the insertional segment is of small size. The case illustrated in Figure 10–3 exemplifies this: a small (0.4% of haploid autosomal length [HAL]) segment from 8q inserted into 10q, with the duplication and deletion outcomes depicted (this case discussed further later). FORM ATIO N O F A QUA DRI VA LE N T
Probably only in exceptional cases, with larger insertional segments, a quadrivalent forms, and this
would enable recombination within the insertional segments. In the review of Van Hemel and Eussen (2000), the mean size of the inserted segment in recombining cases was 1.5% HAL, compared with 1.0% and 0.5% HAL in nonrecombining families in which the imbalances were due, respectively, to duplication and to deletion. With the direct insertion, a recombinant chromosome would be monocentric, and therefore functional. Inverted insertions, on the other hand, would be associated with dicentric or acentric recombinant chromosomes, with the resulting gametes predicted to be nonviable. Consider the large direct insertion depicted in Figures 10–4 and 10–5. Most of the material within the chromosome 5 long arm (q11-q22) has been removed and inserted within the distal long arm of chromosome 1 ( Jalbert et al., 1975). A pachytene configuration at meiosis I such as that depicted would allow for complete synapsis of homologous segments. If no crossover occurred in the insertional loop (and assuming 2:2 disjunction with symmetric
(a) Balanced carrier
8 der(8)
10 der(10)
8
der(8)
10
der(10)
10 der(10)
8
8
10
der(10)
8
der(8)
10
10
(b) Duplication
8
8
(c) Deletion
8
der(8)
10 10
FIGURE 10–3 An insertion from chromosome 8 to chromosome 10, ins(10:8)(q21;q21.2q22), showing (a) the balanced carrier, (b) the duplication, and (c) the deletion states. In this family, the duplication was the only unbalanced form to be observed. (Case of P. A. Bowen; Bowen et al., 1983.) Insertions •
185
d
c a
would be formed, and now three further unbalanced outcomes from symmetric 2:2 disjunction would be possible: gametes [b´,d´], [b´,c], and [a,d´] in Figure 10–4. The duplication/deletion combinations, [b´,c] and [a,d´], are judged to be nonviable, although they might cause miscarriage. The “least imbalanced, least monosomic” combination is the “dup ins” [b´,d´], which leads to a partial trisomy for the insertional segment, 5q11-q22. This was, in fact, the karyotype of the proposita in this family (Fig. 10–5). Actually, this karyotype endows the same genetic imbalance as would the nonrecombinant [b,d] gamete; so in practical terms, it made no difference that this recombination did happen.
b
del 5 5
1 ins 1
Synapsis at meiosis I
b a
SEG M EN T CO N T EN T A N D V I A B I L I TY
c d
b' Gametic outcomes
d'
b'
c
a
d'
del distal 5q dup distal 1q
dup ins
a
c
del ins
dup distal 5q del distal 1q
FIGURE 10–4 Gamete production following formation of a quadrivalent in the interchromosomal insertion, with a single crossover having occurred in the insertion loop. Only one of each sister chromatid is shown. Recombinant chromosomes noted as b´ and d.´ (Based on the case shown in Figure 10–5.)
segregation of centromeres), the same four outcomes noted in the preceding section would eventuate. The gametic combination [a,c] would produce a del(5q11-q22), and the combination [b,d] would produce a duplication for this segment. But if a crossover did occur, two recombinant chromosomes
The viability of the conceptuses—in other words, the risk to the heterozygote of having an abnormal child—depends on the degree of the aneuploid states. Consider the example illustrated in Figure 10–3. A small segment from the middle of chromosome 8 long arm (8q21.2-q22) has been removed and is inserted within the chromosome 10 long arm. This segment comprises about 0.4% of HAL. The heterozygote for this rearrangement could produce two types of unbalanced conceptus: one with a duplication of the segment 8q21.2-q22 (Fig. 10–3b), and one with this segment deleted (Fig. 10–3c). In this family (Fig. 10–6), only the duplication was observed. These individuals had mild to moderate mental retardation and minor physical anomalies (Bowen et al., 1983). A segregation analysis of the family was done, and the segregation ratio was close to 1:1:1:0 for normal:balanced:partial trisomy:partial monosomy. This implies a normal viability for the partially trisomic conceptus, and nonviability for the partially monosomic state. Thus, in this family, the risk for having an aneuploid child is 1/1+1+1+0, or 33%. This assessment is an example of a “private” segregation analysis. A genetically smaller insertional segment has the potential to be viable in both the duplicated and deleted states. For example, Doheny et al. (1997) describe two first cousins, one with a duplication of a segment of 10q, the other with a deletion. The connecting relatives carried an insertion, 46, ins(12;10)(q15;q21.2q22.1).2 The insertional segment, 10q21.2-q22.1, was small, comprising about
2 In the ISCN nomenclature, the recipient chromosome is noted first, followed by the donor chromosome.
186
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
FIGURE 10–5 Interchromosomal insertion with recombinant chromosomes in phenotypically abnormal offspring. Partial karyotypes of 46, ins(1;5)(q32;q11q22) carrier parent (above) and her recombinant child with 46, rec(1)rec(5)dup(5q)ins(1;5)(q32;q11q22) (below). The latter is the [b,’d’] combination in Figure 10–4. The child is trisomic for the segment 5q11-q22. Cartoon karyotype: white, chromosome 1; criss-crosshatched, 5q11-q22; crosshatched, remainder of 5. (Case of P. Jalbert; Jalbert et al., 1975.)
FIGURE 10–6
The pedigree of the family in which the insertion illustrated in Figure 10–3 was segregating.
0.5% HAL. The child with the duplication was identified with learning difficulty in first grade, and her IQ measured at 74; the physical phenotype was rather mild. Her cousin with the deletion had, as an infant, considerable lag in neurodevelopmental progress, which would lead one to anticipate a more serious mental defect when the child is older, and she had a more obviously dysmorphic appearance. An insertion of a very small segment may, on classical cytogenetics, be difficult to detect, although with increasing use of microarray analysis, more such cases will, surely, come to light (Kang et al., 2010). Löffler et al. (2000) were presented with an adult male thought possibly to have fragile X syndrome. In the event, he had an abnormal chromosome 14, with additional material at band 14q13. His retarded brother and normal mother had the same chromosome. Was this an insertion, an
inversion, or what? Fluorescence in situ hybridization (FISH) using microdissection from the abnormal 14 showed a very small hybridizing segment on chromosome 7. Both no. 7 chromosomes in the brothers showed this spot of hybridization, but just one of the mother’s. Going back to the G-banded preparations, and now knowing exactly where to look, a deletion at 7q32-q34 could be discerned on the mother’s other chromosome 7, and the definitive interpretation could be made. She had the karyotype 46, XX, ins(14;7)(q13;q32q34), and the two sons were 46, XY, der(14)ins(14;7)(q13;q32q34)mat. In similar vein, consider the insertion in Figure 10–7, in which two small subbands from 2q (2q33.2 and 33.3) and adjoining parts of q33.1 and q34 are inserted into chromosome 4. This is only about 0.3% of HAL. This rearrangement was at the limit of detection of high-resolution G-banding. In this
2
der(2)
der(4)
4
2
der(2)
der(4)
4
2
2
der(4)
4
2
2
der(4)
4
2
2
der(4)
FIGURE 10–7 A very small insertion, needing fluorescence in situ hybridization (FISH) to be seen clearly. The karyotype of the carrier parent (upper) is 46, inv ins(4;2)(q32; q34q33.1). The child is duplicated for the segment 2q33.1-q34, but this is difficult to appreciate on the G-banded karyotype (middle). FISH with chromosome 2–specific paint (lower) shows the small insertion segment from chromosome 2 present in the der(4). (Case of M. Curtis.) 188
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
family, three of five children had a duplication of the insertion, inheriting from the carrier parent the normal chromosome 2 along with the derivative chromosome 4 containing the insertional segment (2q33.1-q34). The children with this very short duplication had a clinical picture of poor speech development, distractable and aggressive behavior, and subtle facial dysmorphism. Although the range of potential abnormalities may be greater in the case of the heterozygote for a large direct insertion because of the additional risks for gametes having recombinant chromosomes, the outlook is not so discouraging in practice. Often the extent of imbalance associated with a recombinant chromosome is so substantial that nonviability is very likely. In other words, these abnormal pregnancies are lost at a very early stage and do not produce an abnormal child. Each family needs to be assessed individually. The counselor may find it useful to follow the format outlined in Figure 10–4 in making this assessment, in terms of what combinations of imbalanced segments might arise, for the large insertion. The Two-Way Insertion A two-way reciprocal insertion (a rare observation) has the potential for two different imbalances: a partial monosomy from the segment of one chromosome with the reciprocal partial trisomy of the segment from the other chromosome; or the opposite, with a partial trisomy of one chromosome and a partial monosomy of the other (Fig. 10–8). Instructive Cases An insertional translocation involving the critical region for Down syndrome provides an interesting illustration that this small segment is indeed sufficient to produce the phenotype. Lee et al. (2005) describe a father with 46, XY, ins(4;21)(q21;q22.13q22.2), who had a child with typical Down syndrome, having inherited the paternal ins(4) with the small 21q segment, along with two normal chromosomes 21. This case had actually been diagnosed at amniocentesis, when a FISH probe recognizing 21q22 showed three signals; interpretation of the karyotype was a rather more subtle exercise, since 4q21 and 21q22 have similar staining properties, and the small inserted segment thus did not stand out. Because nonreciprocal insertional translocations lead to single segmental imbalances, they can be helpful in delineating genes or phenotypes. Such is the case of an ins(13;11)(q14.1;p11.2p12) segregating
in a family, in which the deletion individuals had biparietal foramina (skull bone deficiencies), multiple exostoses (bone growths), and developmental delay (Shaffer et al., 1993). The description of this family led to the recognition of other deletion individuals, and eventually to the discovery of genes involved in multiple exostoses and biparietal foramina (Wakui et al., 2005; and see Potocki-Shaffer syndrome, p. 318). Gametogenesis Studies Gametic analysis has been reported in two insertion heterozygotes. Goldman and Hultén (1992) examined testicular material from an ins(6;7) heterozygote and demonstrated independent synapsis of the chromosome 6 and chromosome 7 homologous pairs at diakinesis, with the two bivalents occupying quite separate parts of the nucleus. This is a direct demonstration that the segregation scenario set out in Figure 10–2 does happen. Testicular tissue and sperm was studied from one ins(3;10) carrier in whom a very small segment of chromosome 10 (p13-p14) was inserted into chromosome 3 at q13.2 (Goldman et al., 1992). In meiosis I, the pairing chromosomes did not loop out the nonhomologous segments; but in fact the normal chromosome 3 appeared to pair fully with the der(3), and likewise the chromosome 10 and the der(10). This may be heterosynapsis. Sperm karyotyping showed, as expected from the theoretical considerations noted earlier, similar proportions of gametes with normal, balanced, duplication, and deletion chromosomes: the actual figures were 22%, 32%, 24%, and 22%, respectively. No recombinant forms were seen. Possibly, small insertions may show similar meiotic behavior, with absence of looping out and no quadrivalent formation. Spermatogenesis may be compromised in some carriers, a conclusion drawn from the observation that only half as many index cases have carrier fathers as they do carrier mothers (Van Hemel and Eussen, 2000). RA R E CO M PL EXI T I ES
Most nucleolar organizing region (NOR) translocations are harmless (see p. 262). But a NOR insertion into the X chromosome associated with a familial X-linked spastic paraplegia (a condition in which there is stiffness and weakness of the lower limbs, due to neurological deficit at the level of motor neurons in the spinal cord) was apparently pathogenic (Tamagaki et al., 2000). NOR material comprises Insertions •
189
(a) Partial monosomy
{
}
+
Partial trisomy
(b)
Monosomy Trisomy +
Trisomy Monosomy
FIGURE 10–8 Insertional translocations and abnormal segregation. (a) One-way, single segment, or nonreciprocal insertion. The hatched bar represents the portion of the chromosome inserted from the gray chromosome to the white chromosome. Upon fertilization by a normal sperm (region of interest from the gray chromosome is shown), there are two possible outcomes (arrows). The upper set of chromosomes shows the outcome after fertilization of an oöcyte carrying a deleted gray chromosome and a normal white chromosome; this leads to a partial monosomy (deletion). The lower set shows the outcome after fertilization of an oöcyte carrying two normal gray chromosomes and a white chromosome with the insertion; this leads to a partial trisomy (duplication). (b) Two-way, double-segment, or reciprocal insertion. Upon fertilization by a normal sperm, the upper set of chromosomes shows the outcome after fertilization of an oöcyte carrying only the insertion from the white chromosome. The result is trisomy for the white segment and monosomy for the gray segment. The lower set shows the reciprocal product, with trisomy for the gray segment and monosomy for the white segment.
DNA coding for ribosomal RNA. Two brothers and their maternal uncle had the disease, and the carrier mother was unaffected. Plausibly, the inserted material disrupts a “spinal motor neuron gene” in this region, at Xq11.2, and the male hemizygote, with no gene product being made, thus develops the disorder. It cannot yet be excluded that there is an X-linked mendelian disorder whose locus resides in Xq11.2, cosegregating in the family by chance, and the NOR insertion is simply serving as a cytogenetic marker. The discovery of the gene would prove the point. There can be a link with cancer if a tumor suppressor gene is located in the insertional segment (Barber et al., 1994). An extraordinary case is seen in a father who had had Wilms tumor as a child, and whose daughter had retinoblastoma, due to an insertion that was apparently balanced in him, and unbalanced in his child. A segment from 13q14 including the retinoblastoma (RB) gene was inserted into 11p13, this being the site on chromosome 11 of the WT1 Wilms’ tumor locus (Punnett et al., 2003).
GENETIC COUNSELING Insertions are among rearrangements implying the highest reproductive risk. Pooled data from a number of insertion families (Van Hemel and Eussen, 2000) indicate an average risk of having an abnormal child of 32% for the male carrier, and 36% for the female. It may reach 50%. The risk is greater in the small-segment insertion, and smaller in the large-segment. Offering prenatal testing should, in most cases, be the rule. Of the phenotypically normal offspring, approximately half will have normal chromosomes, and half will be insertion heterozygotes. A more detailed discussion follows. S HORT IN SERTIO N SEGM E NT
For the short insertion (say, <1% HAL), the segregation ratio at conception would be expected to be 1:1:1:1 for normal:balanced:duplication:deletion (as discussed earlier). If the insertional segment is not only short but also genetically “small,” both trisomically and monosomically, the maximum risk of having a liveborn aneuploid child would approach 50% (1+1/1+1+1+1). The segment 18q11-q21 (HAL = 0.8%), for example, meets these criteria, as seen in the insertion family presented in Chudley et al. (1974). Carriers for this insertion had all four karyotypic classes of offspring—insertion heterozygotes,
karyotypically normal individuals, individuals with a duplication of a small segment of 18q, and individuals with the same segment deleted—in approximately equal numbers. A similar scenario is presented in Marinescu et al. (1999b), with a family segregating an insertion ins(16;5)(q22; p14p15.3). Here, the “small” segment comprised 5p14-p15.3. In two generations from a heterozygous grandparent, there were two children with 5p–, two with 5p+, four normals, and three carriers. The same level of risk, with a 1:1:1:1 segregation as earlier, is also likely to apply to the very small insertion that requires microarray for its recognition. If viability is reduced or impossible for the trisomic or monosomic conceptuses, the risk would be correspondingly less. Trisomic lethality presumably increases with an increasing fraction of HAL, with monosomic imbalances being more lethal. It may not be possible to make a clear judgment, based on the literature, about the qualitative content of the imbalance, because the insertion involves an interstitial segment of chromosome, whereas most data on record relate to distal segments. A review of the insertional data on record up to 2000, taken from nearly 90 families, is provided in Van Hemel and Eussen (2000), and Figure 10–9 is taken from their paper. Any insertion involving the same open bar (deletion) or filled bar (duplication) segment, or part thereof, will have a significant risk. Schinzel’s (2001) cytogenetic database and the Internet sources ECARUCA and UKCAD (p. 309) may also be consulted; some more distal insertional segments may well be bounded within terminal duplications and deletions that have been described in liveborn individuals. Of course, any unbalanced child in the counselee’s family will provide proof of viability, and an illustration of that particular phenotype. A study of the wider family may provide a guide to the recurrence risk—a “private” segregation analysis, as illustrated earlier in the “Biology” section. But in any case, the starting point with a patient having a short insertion is that the risk for an abnormal child is high, by which we mean in the range 10%–50%. LON G ER I N SERT I O N SEG M EN T
For the direct insertion involving a longer segment (say, >1.5% HAL), there is theoretically an additional risk for the formation of recombinant duplication and deletion chromosomes. But in fact the deletion for a long segment (whether the result of a nonrecombinant or recombinant chromosome) Insertions •
191
FIGURE 10–9 (Above and opposite) Presentation of chromosome segments in which recombinant imbalances have been recorded (on classical cytogenetics), in the child of a parent heterozygous for an interchromosomal insertion. Segments seen only as duplications are shown in filled bars, those seen only as deletions in open bars, and filled and open bars connected show segments observed in either state. Insertions seen only in the balanced state are identified with striped bars. (From Van Hemel and Eussen, 2000, Interchromosomal insertions. Identification of five cases and a review, Human Genetics 107:415–432. Courtesy J. O. Van Hemel; with the permission of Springer-Verlag.)
FIGURE 10–9
(Continued)
would usually impose a nonviable degree of partial monosomy. The dup/del combinations (see Fig. 10–4) are even more unbalanced, leading to spontaneous abortion. Thus, only the duplication (whether nonrecombinant or recombinant) is likely to allow for viability. In the great majority of cases, therefore, the segregation ratio for pregnancies going to term is
1:1:x:0 for normal:balanced:partial trisomy:other imbalances, where x is less than 1, and probably very much less than 1. In the family of Jalbert et al. (1975) discussed earlier (Fig. 10–5), the insertional segment (5q11q22) comprised 2.2% HAL, and this duplication did allow survival, although the child was dysmorphic Insertions •
193
and severely mentally retarded. This case is the sole example of dup(5)(q11-q22) in Schinzel’s database; this specific segment is not listed in ECARUCA nor in UKCAD. The risk for recurrence in this family, or occurrence in another family, must surely be small, and perhaps x is only a low single-digit number. In a family such as that in Abuelo et al. (1988), with an insertional segment comprising most of 3p (p26p13, 2.5% HAL), one could be rather confident that any imbalanced conception would miscarry. The closest viable segment in Schinzel’s database is 3pter-p14, and there are only two cases of this listed; in ECARUCA, the closest match is 3p26-p21.3, a single case. A risk of “almost 0%” for an abnormal child could be offered. Prenatal diagnosis in cases judged to be of this very low risk category would be discretionary; a normal ultrasonographic fetal anatomy scan would likely be considerably reassuring in itself.
in Figure 10–10. These insertions can be within-arm or between-arm, and direct or inverted, and they may undergo incomplete or complete synapsis. These differences can have practical consequences, and we need to consider each in turn. WI T H I N - A R M I N SERT I O N
A shift of chromatin within the same arm is called, logically enough, a within-arm3 insertion. Since both segments shift, essentially switching positions, each could be called an “inserted segment.” If both segments maintain the same orientation toward the centromere, it is a direct insertion. If the orientation of one segment is reversed, it is an inverted insertion. In the case of the inverted inversion, we can distinguish one segment from the other by referring to respective inverted and noninverted segments. In the direct insertion, the shorter of the two segments
I NT E R MED IATE LEN GT H SEGM E NT
Intermediate length segments (1%–1.5% HAL) might imply a risk in the range 5%–10%. But each segment needs to be judged on its merits, both according to the reproductive history in the family, and with reference to the cytogenetic databases. Preimplantation Genetic Diagnosis Preimplantation genetic diagnosis in the context of an insertion presents challenges. If the theory of quadrivalent formation, with possible crossing-over, is correct, the use of additional probes would be necessary to account for this (Melotte et al., 2004).
THE INTRACHROMOSOMAL INSERTION BIOLOGY Intrachromosomal insertions, also known as “centromere shifts,” are very rare, with only about 40 cases published (Madan and Menko, 1992; Ardalan et al., 2005). The cytogenetic recognition can be difficult, with some having originally been interpreted as paracentric inversions with unbalanced meiotic products (Madan and Nieuwint, 2002). The formation of the intrachromosomal insertion is outlined
FIGURE 10–10 The formation of the intrachromosomal insertion. Left, the within-arm insertion, with the inserted segments crosshatched. The normal chromosome is on the left, and the insertion chromosome on the right. Right, the between-arm insertion, with the inserted segment in black. The normal chromosome is on the left, and the insertion chromosome on the right. Compare with the ins(5) shown in Figure 10–15 and the ins(5) in Figure 10–16, respectively.
3 Also called intra-arm, and paracentric insertion.
194
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
can be arbitrarily labeled as the inserted segment, and the longer as the “noninserted” or “interstitial” segment (Madan and Menko, 1992; Barber et al., 1994); since they are both really insertion segments, we can also speak of the “shorter inserted” and the “longer inserted” segments. Array comparative genome hybridization (CGH) enables, in the unbalanced case, a subtler discernment of the inserted segment (Burnside et al., 2009). BE T WEEN - ARM IN SERT I ON
The other type is the between-arm4 insertion, with a segment of chromatin from one arm inserted into a point in the other arm (Fig. 10–11). If we consider the part of the chromosome containing the centromere as the fixed reference point of a chromosome, we can regard the centromeric segment as “staying still,” while the insertion segment shifts from one arm to the other. This somewhat arbitrary point of view allows us to use the term “inserted segment” unambiguously, in the context of the between-arm insertion. Thus, in Figure 10–11, the segment shown in black has moved “up” from the long arm and is inserted into the short arm (rather than the segment containing the centromere moving “down” into the long arm).5
FIGURE 10–11 Gamete production following a recombination between the sites of rearrangement in the between-arm intrachromosomal insertion. At the top of the figure, the normal chromosome is on the left, and the insertion chromosome on the right. There is incomplete synapsis, with ballooning out. (Based on the ins(5) shown in Figure 10–16.)
I NCOMP LETE SY N APSI S
Meiosis perforce proceeds in a modified fashion. Consider the between-arm shift. In most cases, perhaps, the inserted segments fold out so as to allow a good degree of synapsis of the bivalent. This synapsis would include that part of the chromosome between the two inserted segments, that is to say, the centromeric segment. There would be no difference, at least in theory, if the insertion is direct or inverted. One (or any odd number) crossover within the centromeric segment will produce recombinant chromosomes: one with a duplication of the insertion segment, and the other with a deletion (Fig. 10–11). The centromeric segment may be quite long, as a proportion of the whole chromosome, and provide considerable opportunity for crossover. Thus, the genetic risk is expected to be high; and in theory could approach 50%. In other words, the
segregation ratio for the four possible segregant outcomes of normal:balanced insertion:duplication: deletion would be close to 1:1:1:1. According to the level of in utero genetic compromise imposed by the deleted and duplicated states, respectively, the risk for an abnormal outcome in a liveborn child may be correspondingly less. The family illustrated in Figure 10–12 has an intrachromosomal insertion involving the “Potocki-Shaffer segment” (11p11.2), small enough that microarray is needed for its demonstration. With likely no reduced viability of either imbalanced state in utero, the risk to the carrier here may indeed be 50%. The within-arm shift, in the case of the direct insertion, can have a similar folding out of one inserted segment, and its homolog on the normal
4 Also called inter-arm, and pericentric insertion. 5 It may be more complex than this: Wang et al. (2010a) propose that an inserted segment might invert within itself, as a four-break, rather than a three-break rearrangement.
Insertions •
195
(a) I
II Deletion 11p (Potocki-Shaffer Syndrome)
III
N
N
N
Duplication 11p Balanced insertion 11 Normal 11s
N
N
(b) p12 p11.2 q23.1
11
ins (11)
del (11)
dup (11)
FIGURE 10–12 Family tree (a) showing segregation of an intrachromosomal insertion ins(11) (q23.1p11.2p12), with both deletion and duplication observed in the family, and (b) cartoon karyotype to show the nature of the rearrangement. The insertion was detected on BAC microarray-based CGH, and the insertional segment is of approximately 2 Mb in length. Half-filled symbol, balanced carrier; filled symbol, 11p deletion (Potocki-Shaffer syndrome); crosshatched symbol, 11p duplication. The formal karyotypes of the deletion and duplication states are rec(11)del or dup(11)(p11.2p12)ins(11)(q23.1p11.2p12). (Case of J. Gastier-Foster and C. Astbury.)
chromosome, to enable synapsis of the other inserted segment and its homologous region. In Figure 10–13, we depict the shorter insertion segment folded out, with synapsis of the larger inserted segment (it could have been drawn the other way around). Recombination within the larger segment will lead, respectively, to duplication and deletion of the shorter segment in the recombinant products passed on to the two resulting gametocytes. Vice versa, if there is synapsis of the shorter inserted segments, followed by recombination, there would be duplication of the larger inserted segment in one gametocyte, and deletion of this segment in the other. In theory, the longer the larger segment is, the more likely it is that recombination will happen; but nevertheless, cases are on record of crossing-over taking place in very short inserted segments, in both direct and inverted insertions (Webb et al., 1988; 196
•
Rethoré et al., 1989; Barber et al., 1994). Theoretically, if this process were to happen with an inverted insertion, dicentric and acentric products, almost certainly nonviable, would result. COMPL ET E SYN A PS I S , DI R ECT I N SERT I O N
Alternatively, and likely only in the case of the inserted segment being of large size, complete synapsis may be achieved. The insertion and the centromeric segments (between-arm shift) or the two insertion segments (within-arm shift), and their matching segments on the normal homolog, would need to loop back and forth into each other, forming a double loop (Fig. 10–14). Various outcomes are possible from crossing-overs within one or other loop. Considering the direct between-arm shift,
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
from incomplete synapsis with folding out of one of the segments. Crossing-over within the longer inserted segment will lead to recombinant chromosomes deficient or duplicated for the shorter inserted segment (Fig. 10–14i, j). Vice versa, crossing-over within the shorter inserted segment will lead to recombinant chromosomes deficient or duplicated for the longer inserted segment (Fig. 10–14k, l). We illustrate such a case from Webb et al. (1988) in Figure 10–15; equally, this outcome could have arisen from incomplete synapsis, with the longer segments folded out. COM PL ET E SYN A PS I S , I N V ERT ED I N SERT I O N
FIGURE 10–13 Gamete production following a recombination within one of the insertion segments (the longer segments) of a direct within-arm intrachromosomal insertion. There is incomplete synapsis. There are four possible gametic outcomes. Compare with the ins(5) shown in Figure 10–15, although note the subtle difference that in the latter the recombination took place between the shorter inserted segments.
crossing-over within the centromeric segment will lead to recombinant chromosomes deficient or duplicated for the inserted segment (Fig. 10–14a, b). If, however, following complete synapsis, there is crossing-over within the inserted segment, this will lead to the generation of new recombinant forms: chromosomes that are duplicated for terminal p and deleted for terminal q, or vice versa (Fig. 10–14c, d). A notable such example is illustrated in Ardalan et al. (2005), concerning a mother who carried a dir ins(20)(p13q11.21q13.33) (initially thought to be a pericentric inversion). The “shifted” segment was relatively large, about half the length of the chromosome, and the del qter/dup pter recombinant karyotype conveyed a survivable imbalance. If complete synapsis is achieved in the direct within-arm shift, there is no new category of recombinant form beyond the four that could be generated
Recombination in the inverted between-arm insertion, in the setting of complete synapsis, has the same consequences as for the direct insertion as discussed earlier, when crossovers take place within the centromeric segment (Fig. 10–14e, f ). The family illustrated in Figure 10–16 demonstrates this. The recombinant child with a dup(5) could equally have arisen from recombination in a partial synapsis (Fig. 10–11) or in a complete synapsis (Fig. 10–14f), but in either event the crossover is within the centromeric segment. The duplication comprises the inverted insertion segment. If, however, the crossover is in the inserted segment, dicentric and acentric products will result, and, if a zygote were to result from such a gamete, the compromised conceptus will probably degenerate very early and may not even implant (Fig. 10–14g, h). The same fate awaits conceptions from crossovers in the inverted within-arm shift, if crossing-over happens within the inverted segment (Fig. 10–14o, p). If crossing-over is in the noninverted segment, we see the same imbalances (Fig. 10–14m, n) as in the direct within-arm shift (Fig. 10–14i, j). Thus, Rethoré et al. (1989) describe a child with a duplication for the very short segment 5p13.32-p14.2 due to a parental inv ins(5)(p13.31p14.3p15.12) with recombination in the even shorter segment p14.3-p15.11, reflecting the scenario set out in either Figure 10–14n, or in Figure 10–13. A notable example of a three-generational inverted within-arm insertion is the inv ins(15) (q15q13q11.2) family described in Collinson et al. (2004). The grandmother, her son and her daughter, and one grandchild were heterozygous for the insertion, with the detailed karyotype written inv ins(15) (pter → q11.2::q13 → q15::q13 → q11.2::q15 → qter). Three grandchildren were abnormal: one with Insertions •
197
FIGURE 10–14 The range of possible recombinants from crossing-over in one or other insertion loop following complete synapsis of the intrachromosomal insertion. The four panels show, from above down, the direct between-arm insertion, the inverted between-arm insertion, the direct within-arm insertion, and the inverted within-arm insertion. In the loop diagrams, the dots signify the centromere, and the × shows the point of crossover. The insertion segment DE is shown in thick line in the loop and in the recombinant chromosomes. Circled letters provide reference points for text comments. (Adapted from Madan and Menko, 1992.)
Mother
5
ins(5)
5
ins(5)
Child
The reader may have discerned a pattern in the aforementioned construction. Whichever segment recombination takes place in (the active segment, so to say), it is the other (passive) segment that comes to be duplicated or deleted. This is logical. A crossover will create a new version of the active segment that contains a portion from each contributing chromosome—but it will be the same length as it was before. The other, non-crossing-over segments follow, as it were, passively along. COM PL ET E SYN A PS I S , I N SERT I O N SEGM EN TS I N B OT H D I R ECT I O N S
5
rec(5)
5
rec(5)
FIGURE 10–15 Recombination from a direct within-arm shift. Partial karyotypes of an insertion heterozygote mother, and her recombinant child. The karyotypes are 46, dir ins(5)(p14.1p14.3p15.1), and 46, rec(5)dup(5p)dir ins(5)(p14.1p14.3p15.1)mat.6 The child is duplicated for 5p14.3-p15.1, shown as the larger crosshatched segment. The recombination may have arisen from crossing-over within band p14.1 (smaller crosshatched segment) at either partial synapsis with ballooning out of segments p14.3-p15.1, as in Figure 10–13, or from complete synapsis following double-loop formation, as in Figure 10–14k. (Case of L. E. Voullaire; Webb et al., 1988.)
Prader-Willi syndrome (PWS), one with the dup(15)(q11-q13) syndrome (p. 329), and the third with Angelman syndrome (AS). As the reader may already have guessed, the AS grandchild was born to the carrier daughter, while the PWS grandchild was fathered by her son: these two grandchildren had each inherited a deletional rearrangement. The grandchild with the dup(15)(q11-q13) syndrome, 46,XX,rec(15)dup(15)(q13-q11.2)ins(15) (q15q13q11.2)mat, was the carrier daughter’s child, having inherited a duplicational rearrangement; the maternal origin of the duplication was the reason, presumably, for that grandchild’s abnormality. The rearrangements would have arisen following either the scenario set out in Figure 10–13 or Figure 10–14m (the deletion) or 10–14n (the duplication).
If the insertional segment is itself rearranged, such that one part is in direct orientation, and the other inverted, this is an “intrasegmental double inversion,” and represents a four-break, rather than a three-break rearrangement (Wang et al., 2010a); in which case, Figure 10–14c would need to redrawn, following Figure 2 in Wang et al. This reinterpreted crossing-over configuration might allow a closer coming-together of the two segments in the normal and the insertional homologs, and possibly be the basis of a particularly high risk of producing imbalanced gametes. Concerning the X chromosome, if the “critical region” of the X is involved, an insertion may, in spite of being balanced, produce gonadal dysfunction in the female (Grass et al., 1981).
GENETIC COUNSELING The risk to have an abnormal recombinant child, in the 27 families reviewed by Madan and Menko (1992), was 15%, although they considered this quite possibly to be an underestimate. This is an average figure, and it was derived from families studied with classical cytogenetics. We may presume a range from near 50% to zero in the individual case. A high risk is likely if one of the segments is small, and the other long, so that (1) there is a high survivability in both the duplicated and deleted state for the small segment, and (2) with one long segment, recombination may be more likely to take place. In this situation, a figure of 30%–40% may be the appropriate one to offer. Given that the partial aneuploid states will involve interstitial regions of the
6 This karyotype stretches the limits of the short nomenclature, since “dup p” could refer to either 5p14.1 or 5p14.3–15.1. The full nomenclature describes the rearrangement: 46, XX,–5,+rec(5)(pter-p14.1::p15.1-p14.3::p13.3-qter),dir ins(5)(p14.1p14.3p15.1)mat.
Insertions •
199
Mother
5
ins(5)
5
ins(5)
5
rec(5)
5
rec(5)
Child
FIGURE 10–16 Recombination from an inverted between-arm shift. Partial karyotypes of an insertion heterozygote mother and her recombinant child. The karyotypes are 46, inv ins(5)(p13q22q33), and 46, rec(5)dup(5q)inv ins(5)(p13q22q33)mat. The child is duplicated for 5q22-q33 (indicated by the crosshatched segment). The recombination may have arisen from crossing-over anywhere between 5p13 and 5q22 at either partial synapsis with ballooning out of segments 5q22-q33, as in Figure 10–13, or from complete synapsis following double loop formation, as in Figure 10–14f. (Case of N. J. Martin; Martin et al., 1985.)
200
•
chromosome, very little data, quite possibly none, may be on record for the viability and phenotype of the particular segment (but of course the appropriate databases should be checked); and an educated assessment will have to be made. In the case of very small insertions, detectable at the level of microarray, the risk is likely to be at the upper end of the range. Risks are presumably less, and possibly zero, if both segments are long (that is, no recombinants are viable). The risks may also be less—say, below 10%—if both segments are short, which might weigh against recombination; but we have no firm data to buttress this suggestion. As always, a “private” segregation analysis, if the family offers that opportunity, may provide the best estimate of risk. For one specific insertion, Allderdice et al. (1983) calculated a risk of 31% for female inv ins(9)(q22q34.3q34.1) heterozygotes. But prediction is imprecise. One short-segment between-arm shift, 46, dir ins(7) (p22.1p21.4q36.1), with a long centromeric segment for which, from the foregoing, a high risk might have been predicted, in fact produced no liveborn recombinant child in a three-generation family, although some first- and second-trimester pregnancy losses may have been due to unbalanced forms (Farrell and Chow, 1992).
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
11 autosomal ring chromosomes
RING CHROMOSOMES ARE UNCOMMON, and it is even more uncommon for a person with a ring (or someone on their behalf) to seek genetic advice about reproductive possibilities. The typical physical phenotype comprises major dysmorphogenesis and mental retardation, and procreation is not usually a relevant issue. But exceptions exist. Remarkably, some persons with a ring chromosome seem to be of entirely normal phenotype. Only mild mental retardation, or short stature with minor dysmorphism, characterizes some other cases. The ring 20 has a unique association with epilepsy. It is these categories of normal or mildly abnormal phenotype—in other words, of possible reproductive potential—we particularly consider in this chapter, although at the outset we can state that only a few examples of parental transmission of ring chromosomes are known. About 99% of rings arise sporadically (Kosztolányi et al., 1991). The ring X Turner syndrome variant is noted in Chapter 13, and the “tiny ring X syndrome” on p. 484.
BIOLOGY There are two major types of ring chromosome that can be associated either with a normal phenotype, or with a clinical picture of relatively mild mental compromise, growth restriction, and absence of major malformation. First, the full-length or nearly full-length ring that replaces one of the normal homologs with the karyotype 46,(r). Second, the very small ring comprising pericentromeric chromatin, which exists as a supernumerary chromosome, with the karyotype 47,+(r). Individuals with either of these types of ring may have intact fertility and may present with questions about risks to their offspring. A third type of ring, in which the phenotype would always be abnormal, may have a complex structure at the breakpoint junctions with terminal deletions and duplications (Rossi et al., 2008b); we do not further discuss this category. We deal with the first two categories separately and list some reported cases of individual ring chromosomes.
•
201
The Apparently Balanced Ring Chromosome, 46,(r) We may list these theoretical mechanisms that could lead to the generation of a ring that might appear, on classical cytogenetics, to be balanced: (1) Fusion of telomeres, without loss of other chromosomal material (2) Deletion of subtelomeric material at terminal p and/or q arms, with fusion of the exposed ends, with only the repetitive telomeric segments lost (telomere healing) (3) Deletion of “small” amounts of euchromatin at p and/or q arm, with fusion of the exposed ends In the first two scenarios, no genes are lost, and no haplo-insufficiency is imposed. In the third case, and if the deletions included actual genes, and if these genes were dosage sensitive, then there would be a phenotypic effect. Further, the circular structure of the ring of itself would then compromise postzygotic mitotic cell division. The rings would become entangled, broken, doubled, or otherwise disrupted, following sister chromatid exchange during the cell cycle (Fig. 11–1). Thus, daughter cells arise that would be partially or totally aneuploid (whether trisomic or monosomic) for the chromosome in question—“dynamic mosaicism.” These cells might die; some, however, could survive in the mosaic state and presumably make an unfavorable contribution to the phenotype. This continuous generation and loss of cells would seriously undermine the growth rate, although it might not greatly influence the quality of growth. The result would be the “general ring syndrome”—whichever autosome is concerned—of marked growth retardation, mild to moderate cognitive impairment, minor dysmorphogenesis, and, perhaps, intact fertility; intriguingly, café-au-lait macules are quite often seen (Kosztolányi, 1987; Sodré et al., 2010). An alternative view is that genomic imbalance suffices, of itself, to lead to phenotypic abnormality, in some cases, at least; and that there is no need to invoke a general ring syndrome (Rossi et al., 2008a). Zollino et al. (2010b), with specific reference to the r(14), propose that silencing of genes may be contributory. D E TAILS O F MEIOTI C BE HAV I OR
At gametogenesis in the 46,(r) heterozygote the expectation is, other things being equal, for symmetric 202
•
disjunction, with 1:1 segregation of the ring and the normal homolog (Fig. 11–2). Thus, half of the conceptuses would be entirely normal karyotypically, and half would carry the ring. If “dynamic mosaicism” then occurred, these latter may be lethal in utero, or those surviving to term might have phenotypic abnormality. There are tentative grounds for considering that the ring heterozygote might have an increased risk for nondisjunction, resulting in 2:0 segregation. In this event, with respect to chromosomes 13, 18, or 21, a child with the respective trisomy might be born. So far, this is on record only in the case of a child with ring Down syndrome, 47,+r(21), born to a 46,r(21) parent (Kosztolányi et al., 1991). The r(21) parent is also at risk of having a Down syndrome child due to a recombinant duplication 21 (Howell et al., 1984; Fryns and Kleczkowska, 1987; Miller et al., 1987). Almost all instances of parent-to-child ring transmission involve the mother as the carrier parent (MacDermot et al., 1990). Probably, spermatogenesis is compromised in the presence of a ring chromosome, and infertility is the consequence for most male heterozygotes. Most of the autosomes are represented in the list of rings, in the form 46,r(A). In a few there has been an association with phenotypic normality and parenthood, and we provide commentaries later. (As noted earlier, there is sometimes mosaicism, more usually with one cell line monosomic for the chromosome concerned, and occasionally a minor cell line with two copies of the ring.) Ring 1, 46,r(1). Few reports exist (Gardner et al., 1984; Cutenese, 2000). Growth retardation is typical. Ring 2, 46,r(2). Prenatal and postnatal growth retardation and microcephaly are consistent features. We have seen a case of 46,(r)(2)/46,N mosaicism, a profoundly retarded girl, with dramatic levels of dynamic mosaicism: 22% of cells were tetraploid, and most of the derivative rings were in the tetraploid cells (Sutherland and Carter, 1978). Lacassie et al. (1999) summarize eight published cases and provide a photographic record of their own patient from birth to age 10 years, a microcephalic child with some mild cognitive and behavioral compromise, and profoundly growth retarded. Dee et al. (2001) showed a subtle distal 2p deletion in a ring 2 child with a similar phenotype, and they
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Replication
0 SCE
Separation of centromeres
Symmetric segregation
1 SCE
2 SCEs
Attempted separation of centromeres
Double ring
Interlocked rings
Broken double ring
Broken ring
FIGURE 11–1 Dynamic mosaicism. The single-chromatid ring chromosome replicates during interphase. Sister chromatid exchanges (SCEs) may, or may not, take place. At meiosis, if there are no SCEs (left), segregation is symmetric (dotted arrows represent spindles drawing homologs to opposite poles). If there is one SCE, a double-sized ring is generated (middle). With each centromere being tugged to opposite poles at anaphase (dotted arrows), the chromosome may break. If there are two SCEs, in the same “direction of rotation” (right), the two rings become interlocked. Breakage, or other mechanical compromise, is the consequence. A second SCE in the opposite direction of rotation would restore the situation.
suggest that some other cases of r(2) may also have very small deletions. It thus remains an open question whether the phenotype is truly a manifestation of the general ring syndrome, or that it is due, in part at least, to a distal deletion. No parent to child transmission has been recorded.
Ring 4, 46,r(4). Sigurdardottir et al. (1999) describe a growth-retarded infant with normal developmental progress and whorled areas of hyperpigmentation and hypopigmentation. The r(4) was a true telomere-to-telomere fusion, as demonstrated with fluorescence in situ hybridization (FISH) using Autosomal Ring Chromosomes •
203
intellectual deficit (Kaur et al., 2008). Vermeesch et al. (2002) described a patient with microcephaly and height well below the third centile; a few cells had double rings. In this case, they demonstrated a subtelomeric fusion: FISH probes for subtelomeric sequences gave positive staining on the ring chromosome, but no staining occurred with telomere probes.
FIGURE 11–2 Meiosis with symmetric segregation in the ring heterozygote.
subtelomeric probes. If the ring formed following terminal deletions, the individual might or might not present a Wolf-Hirschhorn syndrome phenotype, according to the extent of the deletion into 4p (Balcı et al., 2006; Kim et al., 2009). We have seen a man with 46,XY,r(4) manifesting the “general ring syndrome”: he was considerably shorter than his brothers, and his occupation of warehouse manager compared with the professional qualifications of his siblings. Nevertheless, he could fully appreciate the genetic implications of his condition, and he and his wife chose to have donor insemination. Ring 6, 46,r(6). Urban et al. (2002) reviewed 23 cases. Hydrocephalus was a common observation. At one end of the spectrum, malformations and microcephaly with severe retardation are typical. Kara et al. (2008) describe epilepsy as part of the phenotype in a patient whose r(6) included a 6q deletion. At the other end, a much milder phenotype of growth retardation evokes the general ring syndrome. An example is provided by the case of a young woman with mild dysmorphism and short stature, but normal psychomotor development and intact fertility (her son had a normal karyotype), reported in Höckner et al. (2008). The r(6) had approximately 200 kb deleted from each arm of the chromosome. No instance is known of parent-tochild transmission of a r(6). Ring 7, 46,r(7). In a review of 16 cases of r(7), most had presented with microcephaly and 204
•
Ring 8, 46,r(8). A man having a ring 8 with megabase-size deletions at 8pter and 8qter, and whose intellectual deficit was less marked than in most r(8) cases, proved to be mosaic, with a upd(8) pat 46,XY cell line (Gradek et al., 2006). The probable sequence was as follows: 46,r(8) at conception, the ring of maternal origin; mitotic loss of the ring to give a 45,–8 cell; and subsequent “rescue” of the monosomic line by duplication of the normal (paternal) homolog. This mechanism may have operated in some other ring cases in which there is a concomitant normal cell line. Ring 9, 46,r(9). The phenotype in the r(9) is comparable to that of deletion 9pter and 9qter cases (Purandare et al., 2005; Sheth et al., 2007). Common elements include dysmorphism, microcephaly, cardiac malformations, growth and psychomotor retardation, and skeletal anomalies. A particular feature may be ambiguous genitalia or, sometimes, sex reversal, caused by deletion of DMRT1 on 9p. The karyotype has been reported in a case at prenatal diagnosis, in which subtelomeric 9p and 9q deletions were proven; fetal defects were documented post termination (Chen et al., 2006d). Ring 10, 46,r(10). Gunnarsson et al. (2009) report a girl with growth and psychomotor retardation, microcephaly, congenital heart defects, and dysmorphic features. The nonmosaic ring was characterized by array comparative genome hybridization (CGH) and showed terminal deletions of 12.5 Mb at 10q26.12-qter, and 285 kb at 10p15.3-pter. Ring 11, 46,r(11). A mildly dysmorphic and developmentally delayed child had a ring 11 in which the 11p15.5–11p15.4 segment was duplicated, and although this was of paternal origin, he did not present a Beckwith-Wiedemann phenotype (p. 360), but he did develop bilateral Wilms tumor (Carella et al., 2010). Ring 12, 46,r(12). Parmar et al. (2003) review the findings in six cases of 46,r(12). Growth retardation
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
and intellectual compromise of varying degree were consistent features. In one 46,XY,r(12)(p13q24.3) [85%]/46,XY[15%] mosaic case, a man in his twenties presented with infertility associated with severe oligospermia; the diagnosis led to retrospective review, and it was noted that he had been assessed as a child for delayed learning and microcephaly (Martin et al., 2008). He also had a number of caféau-lait skin macules, misleadingly the basis of a previous diagnosis of neurofibromatosis; but as noted earlier, this sign is observed in a number of ring chromosome syndromes. Ring 13, 46,r(13). The typical phenotype, due to the distal 13q deletion component of the ring, presents microcephaly and poor psychomotor development, and genital malformation (WalczakSztulpa et al., 2008). Bedoyan et al. (2004) report mother-to-daughter transmission of a ring 13 chromosome, in which there had been loss only of subtelomeric material. The mother attended a special school; at age 21, she “showed no difficulties with speech, could read a newspaper, and worked as an assistant in a day-care center.” Her daughter had presented with delayed language development. Ring 14, 46,r(14). This chromosome is prone to ring formation, with over 60 cases reported since one of the earliest reports (Gilgenkrantz et al., 1971). The question of “intact” (i.e., no genes lost) versus deleted ring 14 was addressed in Zollino et al. (2010b), these two types being observed in about one-third and two-thirds, respectively. These authors provide photographs of patients in whom the rings were intact, and with 14q deletions of 0.65 Mb, 1.5 Mb, 2.3 Mb, and 3–5 Mb extent, respectively. No clear genotype-phenotype links could be drawn, albeit that in the least affected child in their series, a child of “low-normal intelligence,” the ring was intact. One distinctive feature is a progressive microcephaly, the head circumference at birth typically being normal. Rare cases may present with Prader-Willi-like features (Tzoufi et al., 2007). Given the fact of chromosome 14 being subject to a parent-of-origin effect (p. 361), it was notable that none of these cases had uniparental disomy. A more severe phenotype was seen in one case diagnosed prenatally, in which skeletal anomalies reminiscent of paternal UPD 14 might have been due to loss of the maternal copy of MEG3 (Quenum-Miraillet et al., 2008). The case in Nucaro et al. (2010) demonstrates the complexity
that some rings may show with high resolution studies such as array-CGH. As for transmission of the ring, Bowser-Riley et al. (1981) described a 46,XX,r(14) mother “at the lower end of the normal range” of intelligence, who had two retarded 46,XX,r(14) daughters (and a third 46,r(14) pregnancy which was terminated). Ring 15, 46,r(15). Parent-to-child transmission of 46,r(15) is recorded (Horigome et al., 1992). Glass et al. (2006) illustrate the variability of the r(15) phenotype, contrasting an infant followed up after prenatal diagnosis, who manifested severe growth retardation, dysmorphism and malformations, and marked developmental delay; and an adult woman, employed in a plant nursery, referred for karyotyping for the possibility of Turner syndrome. The infant had a larger distal 15q deletion, and more complex “dynamic mosaicism,” compared with the adult (but age at sampling, and tissue variability of ring behavior, prevent clear correlations being drawn); and, applying array-CGH, they could distinguish the respective rings as 15q26.2→qter and 15q26.3→qter. The ring with the larger deletion lacked the growth factor receptor gene IGFR1, and this likely contributed to the severe growth restriction in the infant; this association has been reported in a number of other r(15) cases. The power of microarray analysis is further illustrated in Manolakos et al. (2009), a prenatal diagnostic case in which an apparently balanced r(15) was shown at CVS and further defined at amniocentesis as 46,XX,r(15)(p11.1q26.3)[21]/45,XX,−15[9]. MLPA and 15qter FISH showed no imbalance, but on microarray, a ~500 kb deletion (approx. nt 99,831,000–100,168,000) at distal 15qter was identified; the reader might wish to enter these numbers into the UCSC or Ensembl genome browsers, and observe how very distal the deletion is, and how few genes (at most, three) are deleted. The fetus was abnormal. Ring 17, 46,r(17). Ring 17 may present with the severe neurological picture of Miller-Dieker syndrome (p. 322), if the deletion extends to the LIS1 locus, or a not quite so severe phenotype with epilepsy, microcephaly, and mental retardation (and café-au-lait macules), if only subtelomeric 17p sequence is removed (Ricard-Mousnier et al., 2007; Surace et al., 2009). Analogous to the ring 22 (see later), somatic loss of the chromosome can be the “first hit” leading to neurofibromatosis type 1 (Havlovicova et al., 2007). Autosomal Ring Chromosomes •
205
Ring 18, 46,r(18). Consistent features include microcephaly, mental retardation, seizures, maxillofacial dysmorphism, and clefting (Koç et al., 2008; Ono et al., 2010). Stankiewicz et al. (2001a) studied seven phenotypically abnormal cases in some detail. Loss of 18q material was consistent, and thus a picture reminiscent of 18q–resulted, while loss of 18p was variable. The abnormal child in Miller et al. (2003) had two rings, one derived from 18p and the other from 18q. A mosaic parent can be phenotypically normal but have a high risk to have an abnormal r(18) child (Fryns et al., 1992b). Yardin et al. (2001) document the history of a woman with the ring 18 syndrome, her karyotype 46,XX,r(18)(p11.3q23)[32]/45,XX,– 18[4] on peripheral blood analysis. Six pregnancies all had abnormal outcomes, and the three karyotyped also displayed the same mosaicism. Ring 19, 46,r(19). Flejter et al. (1996) describe a normal mother having ring 19 mosaicism, 46,XX/46,XX,r(19), with only 4% of cells (lymphocytes) having the ring, while her abnormal daughter was 46,XX,r(19) in 98% of cells. A telomeric probe hybridized to the ring, suggesting a telomere-to-telomere fusion format; and a small ring would be unlikely to undergo dynamic mosaicism, as discussed earlier. Speevak et al. (2003) report a very similar case. Ring 20, 46,r(20). Two different cytogenetic forms of this syndrome exist. In most, there is r(20)/ normal mosaicism, and a tip-to-tip fusion structure of the ring, of presumed postzygotic origin. In some, there is distal deletion of one or both arms, of likely meiotic origin, and in these cases nonmosaicism is typical, and with a more severe clinical picture (Conlin et al., 2011). Epilepsy is the notable clinical feature (Vignoli et al., 2009). The electroencephalogram (EEG) has a characteristic pattern, with trains of “theta waves.” Any patient with epilepsy who has long runs of epileptiform activity on the EEG in the nonseizing state, which may or may not be associated with confusion or diminished consciousness, should have chromosome analysis with this ring chromosome in mind. Zou et al. (2006) suggest, as one possibility, that silencing of the neuronal channel genes CHRNA4 and KCNQ2, at 20qter, due to a “telomere position effect,” might be the basis of the epilepsy in the tip-to-tip ring. An otherwise unaffected parent with a lower level of mosaicism can have affected children with 206
•
the ring chromosome in higher proportion (Canevini et al., 1998). Herrgård et al. (2007) report a mother with 10% r(20) mosaicism, who had an onset of seizures in her mid-twenties, and who was intellectually normal. Her daughter had epilepsy from age 7, and cognitive capacity fell away in subsequent years; her son was always behind in development, showed poor behavior, and had seizures from age 5. These both had 40% of their cells with the r(20). Ring 21, 46,r(21). The cognitive phenotypes can vary from normal to mild retardation (Gardner et al., 1986b). The male 46,XY,r(21) heterozygote may be subfertile (Dallapiccola et al., 1986). FalikBorenstein et al. (1992) report a three-generation kindred. One 46,XX,r(21) heterozygote had had seven pregnancies with four early miscarriages, one normal son, one son with Down syndrome, and one 46,XX,r(21) daughter, the latter herself having a 46,XX,r(21) daughter. Most karyotyped cells in these individuals were 46,r(21), but a few were 45,– 21, and some had a double-size or multisize rings. Short stature, but normal IQ/development, accompanied the abnormal karyotype in these females; one male heterozygote may have had a low-normal intelligence. In one baby with 46,XY, r(21)(q22p11.2)[34]/45,XY,–21[4]/46,XY[14] diagnosed prenatally, and normal on assessment at 10 months, the father proved to have one cell out of 100 with the ring, which may have reflected a somaticgonadal mosaicism (Papoulidis et al., 2010). Melnyk et al. (1995) discuss the difficulties in counseling, relating to uncertainty of the predicted phenotype, in a three-generation r(21) family in which the (nonmosaic) r(21) persons were of normal appearance and intelligence. A 46,XX,r(21) mother had a prenatal diagnosis that showed one 46,XY twin and the other with 46,XX,r(21)/ 45,XX,–21 mosaicism. Both babies were normal, and the girl’s postnatal karyotype was nonmosaic 46,XX,r(21), the same as in the mother. The 45,XX,–21 cell line on amniotic fluid culture may have been of extra-fetal origin or may have arisen as an in vitro artifact. Mother-daughter transmission is recorded in Bertini et al. (2008a), with each having the same karyotype on blood: 46,XX, r(21)/ 45,XX,–21 [98%, 2%]. In this instance, the rearrangement was due to a subtelomeric 21q deletion of 3.4 Mb, and apparently no dosage-sensitive genes had been lost. In their review, Muroya et al. (2002) note other instances of a r(21) parent having a child
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
with a rea(21), possibly indicative of a susceptibility within the ring chromosome to undergo further rearrangement. Their own case illustrated the reverse circumstance: a normal mother with a rather complex der(21) had a mildly mentally retarded son with 46,XY,r(21) (and 4/100 cells 45,XY,–21). A sperm study on a ring 21 infertile man with an extremely low sperm count, karyotyping 45,XY,– 21[3]/46,XY,r(21)[95]/46,XY[2] on blood and with fairly similar proportions on buccal cells, came up with an interesting result: FISH showed most (92%) of 169 spermatozoa to be normal, 7% with the ring, and 1% disomic with the normal 21 and the ring 21. These authors suggested that the (presumed) small fraction in the gonad of normal spermatogonia were selectively favored at meiosis, leading to the majority of gametes being normal (Hammoud et al., 2009). Ring 22, 46,r(22). A handful of inherited cases are on record (Teyssier and Moreau, 1985; Crusi and Engel, 1986; Wenger et al., 2000). In some, the ring is inherited from a phenotypically normal parent to phenotypically normal offspring; in other cases, one or more of the family members with the ring have mental retardation or other clinical features. In some cases, the parent is mosaic and the child has inherited the ring in a nonmosaic state, which may partially explain parent-offspring differences in phenotype ( Jobanputra et al., 2009). Wenger et al. report an example of parent-to-child transmission. The r(22) mother had required special education at high school. Her son had bowel and heart defects, with very little language development by age 20 months. By a strange coincidence he had, on his other chromosome 22 chromosome, a de novo del(22)(q11.2). Infertility, with complete spermatogenic arrest, is recorded in an otherwise normal man (Zuccarello et al., 2010). Some ring 22s have a more proximal q arm breakpoint and are deleted for the ARSA gene at 22q13; in such cases, the phenotype is essentially that of the 22q13.3 deletion, also known as Phelan-McDermid syndrome (p. 327) (Koç et al., 2009; McGaughran et al., 2010). Denayer et al. (2009) make the point (and nicely illustrate) that café-au-lait macules may be observed in this, and other ring syndromes, and, as noted earlier also with respect to the r(12), that this may mislead to a diagnosis of neurofibromatosis type 1. A ring 22 may, of itself, function as a “first hit” in the generation of tumors of neural crest origin, due
to neurofibromatosis type 2 (NF2). The NF2 gene is located at 22q12. The neural crest is the embryonic tissue that gives rise to (inter alia) the investing membranes of nervous system structures. Due to its mitotic instability, a cell line in this tissue might lose the ring and thus become monosomic for 22. Subsequently, a mutation occurring in the NF2 gene on the remaining intact homolog would be “exposed” and allow a classic tumor, a schwannoma of the eighth cranial nerve or a meningioma of the cranial or spinal meninges, to develop (Denayer et al., 2009).
The Supernumerary Small Ring, 47,+(r) A supernumerary chromosome implies, naturally, a partial trisomy. Daniel and Malafiej (2003) presented six cases of their own and reviewed the literature. Generally, it is only when the ring chromosome is very small, or when there is mosaicism with a substantial fraction of normal cells—in other words, where the overall load of genetic imbalance is small—that a question of genetic risk for offspring of the heterozygote will be relevant. Postnatally ascertained cases have naturally presented with an abnormal phenotype, but a fraction of cases come to attention fortuitously, some being phenotypically normal. Mosaicism complicates the interpretation. A few cases are known in which a parent with low-level mosaicism has had an abnormal child with a higher proportion of the cells with the ring. Furthermore, the levels of mosaicism as determined from a peripheral blood sample may not necessarily reflect the levels in other tissues, and including brain; and in a number of rings, little correlation is recognized between the degree of mosaicism and the severity of phenotype. Small supernumerary rings have been reported for every autosome except chromosome 17, as listed in Daniel and Malafiej (2003). Brief sketches of some of these follow, with particular reference to recorded cases in which a parent with the ring has had offspring. Ring 1, 47,+r(1). Callen et al. (1999) presented a series of patients with very small supernumerary r(1) chromosomes ranging in phenotype from normal to abnormal and showed that the size of the ring was correlated with phenotype. Mother-tochild transmission is documented in Bernardini et al. (2007), and these authors summarize the Autosomal Ring Chromosomes •
207
literature to that time. They propose that clones RP11–110B10 and RP4–646P11, mapping to 1p and 1q, respectively, can serve as useful boundaries, and if the ring includes additional material extending beyond these points, an abnormal phenotype is to be anticipated. Kosztolányi et al. (2011) report a three-generation family, (normal) grandfather to (nearly normal) mother to two (abnormal) children, this appearance of “anticipation” being actually a reflection of ascertainment bias. Prenatal diagnosis of 47,+(1) is reported (Wray et al., 2007). Ring 2, 47,+r(2). A 47,XX,+r(2)/46,XX mother with minor facial dysmorphology and apparently otherwise normal had a son with mosaicism for the same tiny ring chromosome, who presented with mental retardation and a psychotic disorder (Giardino et al., 2002). The ring was present in 54% of cells (peripheral blood) in the mother, and 80% in the son. Ring 3, 47,+r(3). A normal mother and her normal infant son had the karyotype 46/47,+r(3), at frequencies of 33% (mother’s lymphocytes) and 41% (prenatal diagnosis in the son, amniocyte analysis) (Anderlid et al., 2001). Ring 4, 47,+r(4). Bonnet et al (2006) review the ring 4 and describe their own case of a child of low-normal intellect, in whom they demonstrated up to three copies of a very small ring chromosome, about 20 Mb in size, in 82% of cells. Three recorded diagnoses were from amniocentesis; all three pregnancies were terminated, with the very severe brain defect of alobar holoprosencephaly identified in one. In a dramatic display of mosaicism, Soysal et al. (2009) report a case with a 46,XY,r(4)[83]/45,XY,– 4[6]/47,XY,r(4),+r(4)[5]/48,XY,r(4),+r(4),+dic r(4)[1]/46,XY[5] karyotype in a patient with cortical dysgenesis, mild mental retardation, seizures, and hip dislocation. Ring 7, 47,+r(7). Tan-Sindhunata et al. (2000) report a family in which the mother of low-normal intelligence, and two of her three children, had mosaicism for a very small supernumerary ring, 47,+r(7)/46,N. Although the fractions of mosaicism were similar in the three (about 50%), the children were more severely affected, at least with respect to language acquisition, than their mother. Speculatively, this could reflect, in the mother, a lesser “ring load” in the brain. Her other child was normal. Similar 47,+r(7) cases are recorded in the 208
•
reviews of Lichtenbelt et al. (2005) and Bertini et al. (2008b); in two, Silver-Russell syndrome was due to UPD 7. The additional copies of the STX1A and LIMK1 genes, common to many r(7) cases, may contribute importantly to the developmental deficits. Ring 8, 47,+r(8). The size of the r(8) correlates well with phenotypic outcome, and comprehensive molecular evaluations of r(8) chromosomes would facilitate karyotype-phenotype correlations (Bettio et al., 2008). Daniel and Malafiej (2003) report a normal woman karyotyped because she had had a child with Wolf-Hirschhorn syndrome, and who turned out to have a very small r(8) in 27% of lymphocytes. A phenotype suggestive of the MURCS (Müllerian and renal aplasia, cervicothoracic somite dysplasia) association was seen in the patient of Loeffler et al. (2003), a mildly retarded teenage girl, in whom 70% of cells contained a tiny r(8) chromosome. Filges et al. (2008) studied a developmentally delayed girl mosaic for a small (not tiny) ring 8, and applying array-CGH, they could describe the extent and size of the ring, namely, 47,XX,+r(8)(::p11.21→ q21.2::)[18]/46,XX[12], comprising 43.8 Mb. Bettio et al. (2008) document a prenatally diagnosed de novo very small ring comprising about 5 Mb of proximal 8p and 8q euchromatin, in mosaic state (50% of cells with the ring on CVS, 90% at amniocentesis, and 96% at postnatal blood sampling). Although early infant development was within the normal range, by age 3 it was clear that language acquisition was poor, and that behavior was affected. A similar case in Gole and Biswas (2005) concerned prenatal diagnosis (amniocentesis and fetal blood) with 50% mosaicism 47,XY,+r(8) (?p11.2q11.2)/46,XY; the child was developing apparently normally at 9 months. However, the level of mosaicism may not necessarily be a useful determinant in prognosis. Familial transmission is known. A normal father, a university graduate, with low-level mosaicism for a very small supernumerary r(8) had two nonmosaic 47,XX,+r(8) daughters (Rothenmund et al., 1997). They were intellectually handicapped and displayed emotional immaturity, although their physical growth was normal. Ring 10, 47,+r(10). Few prenatal diagnoses are on record. Sung et al. (2009) review three reports and describe their own case of mosaic 47,XX,+ r(10)/46,XX detected at amniocentesis and confirmed in the newborn. They extended the study
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
with array-CGH and could thus describe the imbalance as arr cgh 10p11.2q11.2(CN_519687→ CN_541524)×3. The child was normal on assessment at age 1 year. Ring 12, 47,+r(12). Rings of chromosome 12 may be of variable makeup: 12p material, 12q, both 12p and 12q, or of uncertain components. No clear clinical phenotype has emerged, other than abnormality in all (Davidsson et al., 2008). Yeung et al. (2009b) document a case in which the ring 12 included two copies of 12p, thus determining a Pallister-Killian phenotype (p. 305). Ring 15, 47,+r(15). A exceptional case is that of a small bisatellited supernumerary marker chromosome (SMC) derived from chromosome 15 in grandparent (mosaic) and parent (nonmosaic), evolving into a very small ring 15 in the grandchild. All three, and two other siblings with the SMC, were normal (Adhvaryu et al., 1998). Ring 18, 47,+r(18). Jenderny et al. (1993) describe a phenotypically normal mother with 47,XX,+r(18) in only 2/100 cells on blood analysis, the remainder being 46,XX, and who had a daughter with nonmosaic 47,+r(18). A comparable story is reported in Balci et al. (2011) of a mother with a ring in 10% of cells, whose abnormal son was likely conceived as 47,XY,+r(18), but in whom postzygotic events led to his having the mosaic karyotype 46,XY,r(18)/46,XY. A man with a VACTERL-like clinical picture, and with a normal intellect, carried at low-level mosaicism a r(18) that endowed “octasomy” for a ~5 Mb segment of pericentromeric chromosome 18 (van der Veken et al., 2010). Ring 20, 47,+r(20). Guediche et al. (2010) provide a review of 13 cases, eight ascertained postnatally and five prenatally, with psychomotor and growth retardation as frequent, but not universal observations. Kitsiou-Tzeli et al. (2009) document prenatal diagnosis, following which the child, at age 3 months, was judged to be essentially normal; in contrast, Callier et al. (2009) describe prenatal diagnosis based upon array-CGH, in which the aborted fetus displayed dysmorphic features. These several authors point to the need for precision in the molecular delineation of the ring, to enable a clearer genotype-phenotype correlation (a common plea that could of course apply to any of the ring chromosomes). We know of no adult cases.
Ring 22, 47,+r(22). Mears et al. (1995) document a family in which a phenotypically normal grandfather and father were mosaic for a tiny ring 22 chromosome, 47,XY,+r(22)/48,XY,+r(22),+r(22). A grandchild, also 47,+r(22)/48,+r(22),+r(22) but whose ring chromosomes had increased in size, had cat-eye syndrome (see p. 307). RA R E CO M PL EXI T I ES
Supernumerary Ring with a Balancing Deletion. If a ring chromosome is derived from a segment of chromosome that has been deleted interstitially from an autosome, and if this newly generated ring contains a centromere, it can, in some cases, be transmitted stably at mitosis; and, if so, the karyotype is balanced (p. 158). But the carrier can be at high risk to produce unbalanced gametes. The ring might be transmitted as a supernumerary chromosome, to give a partial trisomy. Or the normal homolog might be replaced by the deleted chromosome, for a partial monosomy. And, even if it is the balanced combination that is present at conception, a substantial risk exists for postzygotic mosaicism (which might or might not generate an abnormal phenotype). If the ring is very small, the balancing deletion may be missed on classical cytogenetics, as Baldwin et al. (2008) describe in a mother whose karyotype, at first sight, was 47,XX,+r(4)/46,XX; but the small r(4) was in fact derived from a deleted segment of 4p on one of her chromosome 4 homologs. Her child, who inherited this small ring, but not the balancing deleted 4, had a “mild speech delay.” Mantzouratou et al. (2009) studied embryos from a couple, the wife being 47,XX,del(22),+r(22), and herself normal. They had had two natural pregnancies, both mosaic 47,+r(22)/46, the first producing an abnormal child, and the second terminated after prenatal diagnosis. Unfortunately, following two preimplantation genetic diagnosis (PGD) cycles, none of the embryos had received the normal, intact maternal chromosome 22, and thus none were transferred. Formation of a Neocentromere. A fragment of a chromosome not containing a centromere would not normally be able to be transmitted during cell division. But if a “neocentromere” is generated (p. 307), its survival may be assured; and again we may be dealing with a supernumerary ring that balances a deletion. Slater et al. (1999) describe such a Autosomal Ring Chromosomes •
209
scenario in an infertile but otherwise normal man. A segment was deleted from one chromosome 1, and this same segment (1p32p36.1) existed as a tiny supernumerary ring chromosome. This man thus has the karyotype 47,XY,del(1)(p32p26.1)+r(1) (p32p36.1). The ring chromosome was able to activate the formation of certain centromere binding proteins, which presumably enabled its stable transmission. A similar circumstance is recorded in Knegt et al. (2003), in this case a phenotypically normal woman who had presented with recurrent miscarriage, and in whom a tiny ring 13 chromosome was derived from an interstitial deletion of the segment 13q21.31-q22.2. Amniocenteses in her fourth and fifth pregnancies demonstrated normal karyotypes. More Than One Ring. Multiple tiny rings were identified in a unique case (Vermeesch et al., 1999). In this retarded male, fibroblasts culture showed four tiny rings per cell, and in lymphocytes, six. Each ring had a functional centromere. The formation of supernumerary rings would be expected to be a sporadic event, unrelated to any particular predisposition. Very rare reports might belie that supposition as a universal truth. Callen et al. (1991) recorded two retarded children each with two supernumerary small rings: one with a r(6) and an r(X), and the other with an r(3) and another small unidentified ring. Shanske et al. (1999b) described monozygous twins both had two very small supernumerary rings, with similar proportions of cells in each child: about 60% with 48,XX,+r(1),+r(16), 30% with either 47,XX,+r(1) or 47,XX,+r(16), and 10% with 46,XX. Although too small to be confirmed as ring chromosomes, Ballif et al. (2007b) reported a patient with two sSMCs, one derived from 11q, and one derived from 17p.
GENETIC COUNSELING Parental Karyotype 46,(r) The great majority of transmitting parents are 46,XX,(r) mothers, presumably reflecting that most male heterozygotes are infertile. The observed risk for the 46,(r) parent to have a child with the same karyotype is a little less than the theoretical 50%, and a figure of about 40% will generally be a fair one to offer. Those offspring inheriting the ring could be expected to present the similar clinical picture as, and indeed quite probably more severely than, their heterozygous parent. In the review of Kosztolányi 210
•
et al. (1991), about one-third of 46,(r) children were more severely affected mentally than their parent. The 46,(r) parent may be an atypical ring carrier, perhaps with a fortunate pattern of mitotic disruption, to have reached the level of social phenotype that procreation would be likely. In the particular case of the 46,r(21) heterozygote, who is often phenotypically normal, there is a small but as yet unquantified risk of having a child with Down syndrome due to an uncommon karyotype: 47,+r(21), 46,rob(21q;21q) or 46,tan dup (21q;21q) (Kosztolányi et al., 1991). If, in prenatal diagnosis for a pregnancy of a r(21) heterozygote parent, the same r(21) karyotype were demonstrated in the fetus, based on the slender evidence thus far available, the chance for phenotypic normality would seem to be “substantial,” but a (probably mild) degree of abnormality can by no means be excluded. As Kennerknecht et al. (1990) comment “accurate phenotype-karyotype correlations cannot be made, since there are carriers with a stable ring chromosome who are affected, whereas others with an unstable ring have a normal phenotype and vice versa.” In a person who is mosaic on somatic analysis, with a 46,N/46,(r) karyotype, the mosaicism might extend also into the gonad. This would convey an important risk to have a nonmosaic 46,(r) child.
Parental Karyotype 47,+(r) Each ring needs to be assessed individually, and careful cytogenetic analysis is urged. Reference to the brief outlines earlier will give a sense of the range of outcomes. A nonmosaic parent with a very small ring might be expected to transmit the abnormal chromosome with 50% probability, assuming (and this may not necessarily be the case) meiotic and mitotic stability. The parental phenotype would, in principle, predict that of the 47,+r child. Mosaicism in the parent, and potential mosaicism in the child, considerably complicate prediction. A higher-grade mosaicism in the child than in the parent, or complete nonmosaicism in the child, would be expected to produce a more severe phenotype, possibly lethal in utero. Parental Karyotype 47,del(A),+r(A). In the ring with a balancing deletion (see earlier), normality in an offspring can only be regarded as secure (other things being equal) in the context of the
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
normal homolog (A)1 having been transmitted from the 47,del(A),+r(A) parent. Even though the carrier parent may be normal, the risk is high that the same balanced karyotype in a conceptus could be followed by postzygotic misdivision, with the eventual
generation of offspring who would be partially trisomic, or partially monosomic, for the autosome concerned, and thus abnormal. A detailed discussion is offered in Mantzouratou et al. (2009).
1 “A” indicates any autosome.
Autosomal Ring Chromosomes •
211
12 complex rearrangements
COMPLEX CHROMOSOMAL REARRANGEMENTS (CCRs) occurring in phenotypically normal persons are rare. Familial cases comprise a minority, with only about 35 examples recorded by the early 2000s (Berend et al., 2002b). Three or more chromosomes are involved, and a considerable variety of rearrangements are possible. Translocation may involve distal segments, as in the usual reciprocal translocation, or interstitial segments, as in the insertion. An inversion and a translocation, for example, may coexist on the same chromosome.
BIOLOGY Three Major Categories of Complex Chromosome Rearrangement Three major categories of CCR are recognized (Kausch et al., 1988). The most common is the
three-way exchange, in which three segments from three chromosomes break off, translocate, and unite (Fig. 12–1). Most three-way CCRs are familial, usually transmitted through the mother; although in one of the largest kindreds on record, showing fivegeneration transmission, three (great)grandfathers must have been CCR heterozygotes (Farrell et al., 1994).1 More complicated, exceptional CCRs encompass a wide theoretical range, but there are not many actual cases. (These two types are also referred to as CCR types I and II.) The simplest CCR is the double two-way exchange, in which there is a coincidence of two separate simple reciprocal translocations. In a sense, the double two-way exchange is not a “true” CCR, and it might as well be described as a double or a multiple rearrangement (Phelan et al., 1990). Similarly, there may be the coincidence of a reciprocal translocation with a Robertsonian translocation, or with an inversion.
1 In the ISCN description of the karyotype, the order of chromosomes in the three-way CCR is as follows: first, the lowest number (or X) chromosome; second, the chromosome that receives a segment from the first; and last, the chromosome donating a segment to the first listed chromosome. Thus, the karyotype for the CCR shown in Figure 12-1 is written 46,XX,t(2;18;11)(q13;q21.1;p15.3).
212
•
recurrent pregnancy loss, and three prenatal cases, De Gregori et al. (2007) showed imbalances in all except one of the abnormals, and one of the normal women. The imbalances comprised microdeletions, in some just a single one, in some two, and in two patients, three and four microdeletions, respectively. DE TA I L S O F M EI OT I C B EH AV I O R 2
2
der(2)
der(2)
11 der(11)
11
der(11)
18
18
der(18)
der(18)
FIGURE 12–1 A three-way complex chromosome rearrangement. Most of 2q is translocated onto 18q; part of 18q is translocated onto 11p; and the tip of 11p is translocated onto 2q. The individual had presented with multiple miscarriages. (From R. J. M. Gardner et al. (1986a), A three way translocation in mother and daughter, Journal of Medical Genetics 23:90. Reproduced with the permission of the British Medical Association.)
An apparently balanced karyotype may be associated with a normal or an abnormal phenotype. If the individual is phenotypically normal, the chromosome rearrangement is assumed to be truly balanced. These cases are often familial. The original CCR in a family typically arises as a single complex event, rather than sequential changes, at a meiosis during male gametogenesis (Grossmann et al., 2010). In the phenotypically abnormal individual, presumably some submicroscopic imbalance or other genetic defect exists, and these cases characteristically involve a de novo chromosome abnormality. Such a case was proven in a CCR studied by Brandt et al. (1997), in a child with tricho-rhinophalangeal syndrome (TRPS; see p. 317). The child had a de novo apparently balanced t(7;13;8) (p21;q21;q24.1), but using fluorescence in situ hybridization (FISH) probes for the TRPS critical region at 8q24.1, a 3 Mb deletion was revealed. In a systematic microarray-based study of 13 abnormal individuals, two normal women who had had
The carrier of a CCR has a risk for an abnormal conception due either to malsegregation of the derivative chromosomes or to the generation of a recombinant chromosome. Malsegregation follows the general principles as set forth for the simple translocation, but naturally the range of unbalanced combinations is greater. For the three-way CCR, the broad categories of malsegregation are 3:3 and 4:2, and (theoretically) 5:1, and 6:0. Recombination, whether producing a balanced or unbalanced karyotype, is rare indeed, and only eight such familial CCRs were recorded in the review of Berend et al. (2002b). In the exceptional CCR, scarcely ever are meiotic recombinants observed, and the family in Gruchy et al. (2010), in which a CCR with five insertional translocations with eight breakpoints was transmitted over three generations without recombinant offspring, is typical in this respect. T HR EE-WAY CO M PL EX CH R O M OS O M E RE A R R A N G EM EN T
At meiosis in the three-way CCR heterozygote, the expectation is that the chromosomes involved in the rearrangement will come together and form a multivalent (Saadallah and Hultén, 1985; Fig. 12–2). Consider how meiosis would proceed in the rcp(2;18;11) translocation illustrated in Figure 12–1. In theory, a hexavalent configuration would allow full synapsis of homologous segments (Fig. 12–3). If disjunction were then symmetric (3:3), up to 20 possible gametic combinations could occur. The two arising from alternate segregation (arrows in Fig. 12–3) would be the only ones to be balanced; the remaining 18 would be unbalanced to a greater or lesser degree. Were asymmetric segregation (4:2, 5:1, 6:0) to occur, a great variety of extremely unbalanced gametes would result. However, it may be that, in some families at least, a tendency to favor symmetric alternate segregation, and a combination of very early lethality of severely unbalanced conceptuses, imply a fair prospect for achieving a normal pregnancy (Walker and Bocian, 1987). Complex Rearrangements
•
213
9q
2p
9p
4q
2q 4p
2p 2q 9p
9q 4q
4p
FIGURE 12–2 The actual appearance of a multivalent at meiosis I. Electronmicrograph of a spermatocyte from a testicular biopsy of a man with a three-way complex chromosomal rearrangement 46,XY,rcp(2;4;9) (p12;q25;p12); line drawing shows component parts of the hexavalent. From N. Saadallah and M. Hultén, 1985, A complex three breakpoint translocation involving chromosomes 2, 4, and 9 identified by meiotic investigations of a human male ascertained for subfertility, Human Genetics 71:312–320. Courtesy MA Hultén, and reproduced with the permission of Springer-Verlag.)
FIGURE 12–3 Diagrammatic representation of the formation of a hexavalent at meiosis in the three-way 2;18;11 translocation depicted in Figure 12–1. The arrows indicate 3:3 alternate segregation. 214
•
An excess of heterozygotes has been noted among the balanced female offspring (Batista et al., 1994). The risk of having a pregnancy that would go to term but produce an abnormal child reflects the nature of the rearrangement—that is, whether there are possible chromosomal combinations that would lead to aneuploidy for a survivable amount of genetic material. Thus, considering the preceding rcp(2;18;11) example, three unbalanced combinations, one 3:3 and two 4:2, might be expected to be viable (Fig. 12–4). Batista et al. (1994), reviewing 29 families with a CCR, determined that an abnormal live birth is most commonly (78%) due to 3:3 adjacent-1 segregation, followed by 4:2 segregation. Recombination would add yet further possibility of imbalance, but this is, as mentioned earlier, very rarely seen. The concept of adjacent-1 and adjacent-2 segregations can be applied in the setting of the CCR, in the case of 3:3 segregations. Thus, the segregant gamete shown at left in Figure 12–4, having one of each chromosome pair represented (one of each
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
FIGURE 12–4 Three segregant outcomes of meiosis in the rcp(2;18;11) heterozygote shown in Figure 12–1, that might be expected to produce viable but unbalanced offspring. The 3:3 adjacent-1 gamete on the left may be the one most likely to be produced.
centromere), would reflect adjacent-1 segregation. An example of 3:3 adjacent-2 segregation is given in Xu et al. (1997). A mother had the karyotype 46,XX,t(5;16;22), and cytogenetic analysis of her morphologically abnormal fetus following intrauterine death at 16 weeks gestation showed 46,XY, der(5),der(16),–22,t(5;16;22). In this case, the abnormal ovum would have had one chromosome 5, two chromosome 16s (one normal, one the derivative), and lacked a chromosome 22. 4:2 segregation particularly characterizes CCRs in which an acrocentric chromosome is a component. Schwinger et al. (1975) reported a mother of two children with typical Down syndrome, who herself had a three-way t(7;21;11) CCR. The affected children had an interchange trisomy 21, in that they had, in addition to the maternal translocation pattern, a second intact chromosome 21. Fuster et al. (1997) give an example of a 4:2 malsegregant, diagnosed at chorionic villus sampling, from a three-way paternal t(2;22;11) CCR. The fetal karyotype was interpreted as 47,–2,der(2),der(22)t(2;22;11)(q13; q11.2;q23). The parents continued the pregnancy, and the retarded and abnormal child had a double partial trisomy: a duplication of the segments 11q23-qter and 22pter-q11.2. The couple had previously had one normal child and three miscarriages. E XCE PTIO N AL CO MPLE X CHROM OS O ME REARRA NGE M E N T
More complex rearrangements imply an even greater potential range of abnormal gametes. Kausch et al. (1988) calculated a minimum of 70 possible unbalanced gametes due to 4:4, 5:3, 6:2, and 7:1 segregations from
an octavalent, in the case of a woman with a fivebreakpoint CCR with translocations of chromosomes 1, 2, 5, and 11 and an inversion of chromosome 1, who had presented with three first-trimester miscarriages. Van der Burgt et al. (1992) report a similarly complex de novo balanced CCR (chromosomes 5, 11, 12, 16; five breakpoints in all) in a mother who had had one miscarriage, one 46,XY child, the index abnormal child, and, as a quite unexpected outcome, a de novo 45,rob(13q14q) at prenatal diagnosis in her fourth pregnancy. One of the most complicated familial CCR scenarios ever described is the case in Röthlisberger et al. (1999). A father carried a de novo rcp(6;7; 18;21)(q22 and q25;q21.3, q31.1 and q32.1;p11.21 and q21.3;q21.3). As this unofficial nomenclature attempts to indicate, there were eight breakpoints altogether, two in chromosomes 6 and 18, three in 7, and one in 21. FISH and spectral karyotyping were needed to clarify the detail of the rearrangement. Most remarkably, among his three children, three different recombinant forms were passed on: a rec(7), a rec(21), and a rec(18). The child with the rec(21) had a balanced karyotype, and he has become a balanced carrier for a simple translocation, 46,t(7;21) (q21.3;q21.3): a “rebuilt” translocation (see later). The other two have partial trisomies for 6q and 7q. A subtler example is the case in Gibson et al. (1997) of a mother with a de novo five-break rearrangement in which two small interstitial segments in 2q and 5q, and two terminal segments in 1q and 5q, exchanged position. Her abnormal child was initially thought to have a possibly unbalanced t(1q;5q), but further analysis revealed his wonderfully complicated true karyotype, and the reader Complex Rearrangements
•
215
may care to draw an ideogram and then compare with the original paper: 46,XY,der(1),der(5),t(1;5;2)(1pter→1q42.3:: 5q23.2 →5qter;5pter→5q21.2:: 2q33→2q35::1q42.3→1qter;2pter→2q33:: 5q21.2→5q23.2::2q35→2qter)mat. An unbalanced CCR could be “corrected” by having the countertype imbalance in another rearrangement. Thus, the mother in Zou et al. (2010) had a three-way CCR t(5;15;7)(q13;q24;p15), which was missing the segment 5q13.1-q14.1. But this segment was otherwise present, as an insertion into a chromosome 4, as der(4)ins(4;5)(q31.3;q13. 1q14.1), thus qualifying as an exceptional CCR. She was phenotypically normal and (unsurprisingly, therefore) balanced on microarray analysis. However, she transmitted this der(4) to her son, whose karyotype was 46,XY,der(4)ins(4;5)(q31.3;q13.1q14.1) mat, and he presented an abnormal clinical picture, due to this segmental duplication. In molecular nomenclature, the imbalance is arr 5q13.1q14.1 (66,783,672–77,559,998)×3 mat, representing a 10.8 Mb duplication. CRYPT IC CO MP LEX CHROM OSOM E RE ARRA N G EMEN T
A CCR may be shown, upon detailed cytomolecular study, to have a greater number of breakpoints or more complex imbalance than had originally been appreciated (Batista et al., 1994; Ballarati et al., 2009); and it may be a little arbitrary as to whether it is categorized as a cryptic or an exceptional CCR. From the days of classical cytogenetics, Wagstaff and Hemann (1995) describe a phenotypically normal father and his two abnormal children, the father and son having an apparently balanced 46,XY,rcp(3;9)(p11;p23) and the daughter apparently 46,XX. On FISH and DNA studies, they could show that the father had a tiny segment of chromatin from the breakpoint in 9p23 removed and inserted into the long arm of a chromosome 8 (Fig. 12–7). At meiosis, it may have been that a quadrivalent formed from the chromosome 3 and chromosome 9 elements, while the two chromosome 8 homologs synapsed independently as a bivalent. On this interpretation, the two children reflect alternate segregation of the chromosome 3 and 9 elements; with respect to the homologs of chromosome 8, the rcp(3;9) son inherited his father’s normal homolog, and so the lack of the 9p23 segment was not corrected, while the 216
•
“46,XX” daughter received the chromosome 8 with the 9p23 insertion. Thus, the son has a del(9)(p23), and the daughter a dup(9)(p23). Further insight may come from the application of newer methodologies. For example, in a study of two previously diagnosed cases of de novo CCR, identified in phenotypically abnormal individuals, de Vree et al. (2009) undertook array comparative genome hybridization (CGH) and M-FISH analyses. In the first case, a teenager, the original karyotype of 46,XY,t(1;18;15)(q32;q21;q24) could be shown to include, as well, a 1.5 Mb microdeletion at 10p13: the full karyotype could now be rewritten (the italics are ours) as 46,XY,der(1)(1pter→1q31:: 10p14→10pter),der(10)(15qter→15q24:: 10p13→10qter) del(10)(p13p13),der(15)(15pter→ 15q24::18q21→18qter),der(18)(18pter→18q21:: 10p13→10p14::1q31→1qter)dn. In the second case, a severely retarded adult, the original karyotype was 46,XY,del(5)(q11),der(11) t(5;11)(q11;q11),der(13)t(11;13)(q11;p11). On modern reanalysis, the rearrangement was seen to be more complex, and the 5q11 deletion not to exist: 46,XY,der(5)(5pter→5p10), der(9)(9pter→9q31:: 5q31→5q31::13q31→13q31:: 9q31→9qter),der(11)(13qter→13q31::5q31→5q10:: 11q10→11qter),dic(11;13)(11pter→11p10:: 13p13→13q31::5q31→5qter)dn. In spite of this complexity, on array-CGH no actual pathogenic genomic imbalance was recognized. Thus, other reasons needed to be invoked (e.g., position effect) as possible causes of the abnormal physical and cognitive phenotype. A familial case in which a supposed simple translocation was shown to be a CCR, and then with a microdeletion coming to light on molecular analysis, is presented in Aboura et al. (2003). A mother and her infant son, the latter with minor dysmorphism and abnormal functional neurology, appeared at first to have the same simple t(3q;22q) translocation. FISH analysis showed this to be a t(3;22;9) (q22;q12;q34.1).Yet finer analysis using a probe to the ABL locus on 9q34.1 revealed a very small deletion at this site on the der(9) of the proband, but not in his mother or in a carrier sister. The deletion was thus presumed to have arisen de novo, during maternal meiosis. These scenarios raise pressing questions: how often might other apparently balanced simple reciprocal translocations have a cryptic
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
complex rearrangement; and how often does a de novo deletion occur on the background of a parental balanced rearrangement (see p. 109)? D OU B LE T WO -WAY COM PLE X CHROM OS O MA L RE A RRA NGE M E N T
Presumably, two separate and independently operating quadrivalents can form (Bowser-Riley et al., 1988). Burns et al. (1986) record sperm karyotypes in a man with a double two-way CCR 46,XY,rcp(5;11) (p13;q23.2),rcp(7;14)(q11.23;q24.1), whose wife had had four miscarriages, a child with cri du chat syndrome, and a normal son carrying the rcp(7;14). Only four of 23 sperm analyzed had an overall balanced complement, and the majority (13) had adjacent-1 segregants for one or the other translocation. Another five showed 3:1 and one sperm showed 4:0 segregation.2 We referred on p. 97 to a couple in which both had a simple reciprocal translocation, and both happened to involve chromosome 7 (7p in one, 7q in the other). It is a useful exercise to imagine how the chromosomes might be transmitted in this family. The couple could, in theory, have a child with a double two-way CCR who would have a combination of their own karyotypes. Providing fertility were not compromised, this child of theirs in
generation II could then, in generation III, have two types of balanced progeny: one with the rcp(7;11), and the other with the rcp(7;22), as in the couple of generation I. We set out this scenario in Figure 12–6. No offspring with a normal karyotype could be produced in generation III, unless recombination between the two der(7) chromosomes were to restore a normal chromosome 7. “Rebuilding” of Chromosomes from a Parental Complex Chromosomal Rearrangement The coming together of several translocation chromosomes during meiosis may set the stage for what Soler et al. (2005) describe as “rebuilding.” The CCR shown in Figure 12–5 with six breakpoints in five chromosomes offers useful illustration (Bass et al., 1985). The woman who carried this rearrangement had four pregnancies, only one of which miscarried, and two produced offspring with a balanced constitution, though different in each child and different from their mother! Recombination involving the centric segment of chromosome 1 led to a daughter receiving a rebuilt der(1), with just the 6p segment being translocated, and a son with a different rebuilt der(1) having just the 7q segment. A son and a grandson had unbalanced karyotypes, which were different, but each led to partial 7q trisomy. Readers who relish esoteric puzzles may wish to
FIGURE 12–5 An extraordinarily complex rearrangement involving three two-way exchanges, with six breakpoints in five chromosomes (see text). (From the family reported in Bass et al., 1985.)
2 This case is instructive in illustrating the point that different rcps can have different meiotic behavior: for example, 60% of the rcp(5;11) segregants but only 30% of the rcp(7;14) showed alternate segregation, the environment accounted for by both translocations acting in the same gonad.
Complex Rearrangements
•
217
Parents
7
der (7) der (11)
11
7'
der (7') der (22) 22
‘Child’
der (7)
der (7')
11
der (11)
22
der (22)
‘Grandchildren’
7
der (7) der (11)
11
7
der (7') der (22) 22
FIGURE 12–6 Theoretical potential pedigree of a couple each of whom carries a simple balanced reciprocal translocation: 46,XY,rcp(7;11)(q22;q23) and 46,XX,rcp(7;22)(p13;q11.2). A child of theirs could have a double two-way complex chromosomal rearrangement combining the two parental karyotypes: 46,rcp(7;11) (q22;q23)rcp(7;22)(p13;q11.2). The original simple translocation karyotypes could be restored in the next generation. The reader can determine how, following one recombination, a 46,N grandchild could be conceived. (Courtesy K. L. Butler.)
refer to the original paper. Rebuilding can lead to a simpler rearrangement. Madan et al. (1997) describe a mother with a familial four-breakpoint t(2;3;8) in which the 2q translocated segment had split with 2q23-q33 going to the der(3) and 2q33-qter to the der(8). Her child’s karyotype was a simple 46,t(2;3) with a rebuilt der(3); the simplification resulted from recombination at maternal meiosis between her der(3) and normal chromosome 2. A similar story is told in Tihy et al. (2005). 218
•
A unique case of rebuilding in sequential generations is given in Zahed et al. (1998). A grandfather had two separate translocations: a simple translocation rcp(1;8)(p31;q21.1) and an insertional translocation ins(9;8)(q34;p23.1pter). Thus, he had two abnormal no. 8 chromosomes, one having a segment from distal 1p attached at 8q21.1, and the other having a deletion at 8p23.1. He had a daughter and a son to each of whom he transmitted a rec(8), the same rec(8) to each, in balanced state. This rec(8)
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
FIGURE 12–7 A cryptic complex chromosome rearrangement (and see text). On the original cytogenetic study, father and son appeared to have the same simple balanced translocation, 46,XY,rcp(3;9) (p11;p23), and the daughter seemed to be 46,XX. DNA and fluorescence in situ hybridization (FISH) studies showed a complex chromosomal rearrangement, in which a tiny segment within 9p23 had been insertionally translocated into 8q in the father. Brackets and dotted lines show translocation of two separate segments from distal 9p across to 3p and to 8q respectively. Thus, both the son and the daughter had an unbalanced complement, the son with a deletion, and the daughter with a duplication, for the 9p23 segment. (From the family reported in Wagstaff and Hemann, 1995.)
had a deletion at its p extremity, and a 1p translocated segment on its q extremity. Presumably, his two abnormal no. 8 chromosomes had recombined in meiosis, at a point somewhere between the p23.1 and q21.1 breakpoints. His daughter in turn had two children, and in each of them she restored, by recombination again in this generation—a rebuilt rebuilding!—the grandpaternal chromosomes: the del(8p) in one child, and the der(8)t(1p;8q) in the other. Both children had an unbalanced state, but different in each. One had a straightforward del(8) (p23.1) karyotype, and thus a partial 8p monosomy. The other had the grandpaternal simple rcp(1;8) and would otherwise have been normal; but in addition she inherited the ins(9), which conferred a partial 8p trisomy. The reader may care to draw the chromosomes of the three generations from this
description and check back to Figure 2 in the original paper. Effect upon Fertility In several complex rearrangements, in the female at least, gametogenesis can accommodate itself to the complexity thrust upon it, and the heterozygote may be fertile and have pregnancies that produce phenotypically normal children. However, the rule of the greater vulnerability of spermatogenesis to chromosomal complexity seems to apply particularly in the situation of the CCR, and the male heterozygote is often sterile due to spermatogenic arrest, or subfertile (Lee et al., 2006; Bartels et al., 2007; Ergul et al., 2009). The involvement of an acrocentric chromosome in the CCR may particularly predispose to this male sterility (Gabriel-Robez et al., 1986). Fertility treatment with intracytoplasmic sperm injection (ICSI) may enable fatherhood ( Joly-Helas et al., 2007). Having made these points, a few CCR men do retain natural fertility (Cai et al., 2001).
GENETIC COUNSELING The male CCR heterozygote who is not otherwise known to be fertile should have a semen analysis to check whether sperm are being produced. If there is oligospermia, IVF will have to be considered, and preimplantation genetic diagnosis (see later discussion) may be appropriate. For the heterozygote (male or female) who is fertile, or for whom fertility can be achieved, a conceptus having either a normal chromosome constitution, or the same balanced CCR as the parent, would be expected to produce a normal child. But a high proportion of conceptions have an unbalanced karyotype. Madan et al. (1997) have determined empiric risk estimates. Overall, the risk for spontaneous abortion is 50%, and the risk for a liveborn abnormal child is 20%. The level of risk is related to the mode of ascertainment— whether through the birth of abnormal infants, multiple miscarriage, male infertility with abnormal spermatogenesis, or fortuitously—and to the family history. If multiple miscarriages have been the pattern in the family in the past, it is likely to continue to be so. In such cases, it may be that all unbalanced forms would lead to miscarriage (Creasy, 1989). If abnormal infants have been born, carriers are likely to have a high risk for the same unfortunate event to happen again. For the three-way CCR, it is generally justifiable to advise that, sooner or later, a normal outcome could possibly be expected. Thus, the couple may be Complex Rearrangements
•
219
willing to make continued attempts until a successful pregnancy is achieved. As always, the pedigree should be studied, in order to understand what might be the particular pattern of meiotic behavior with that CCR. If the reproductive history is very unpromising, optimism may need to be guarded, and the reality of a low chance for a normal child faced (Evans et al., 1984). As for the exceptional CCR, the likelihood for a successful pregnancy would be less, and possibly very small. Bowser-Riley et al. (1988) review the specific case of the double two-way translocation and propose that the risk to have an abnormal child would be approximately the sum of the figures derived separately for each rcp. They acknowledge that might be an overestimate due to nonviability of doubly imbalanced combinations, albeit each on its own might be viable.
normal. But this fortunate outcome could not have been “guaranteed.” With an exceptional CCR, interpretation may be aided by array-CGH. Malvestiti et al. (2010) identified a CCR at amniocentesis, following the discovery of multiple fetal malformations at ultrasonography (which naturally gave a strong indication that the karyotype would be unbalanced). The fetal karyotype was interpreted to be 46,XY,der (4)ins(1;4)(q25;q25q31.1), due to a maternal CCR 46,XX, der(1)ins(1;4)(q25;q25q31.1)t(1;5) (q41;q35),der 4)ins(1;4),der(5)t(1;5). Proceeding to array-CGH, the (normal) mother’s genome was balanced, but a fetal deletion at 4q27q31.23, a gene-rich region of about 30 Mb, could clearly be appreciated. Prenatal detection of a de novo CCR is discussed on p. 468. PREI M PL A N TAT I O N G EN ET I C D I A G N OS I S
P RE NATAL D IAG N OSI S
Once a pregnancy is actually achieved, some may prefer initially to rely on first-trimester ultrasonography, declining chorionic villus sampling, and leaving early abortion to happen naturally if that would be the case, as an unfortunate previous miscarriage history might well cause a heightened sensitivity to the small risk associated with prenatal diagnosis. Others may prefer the early information that a chorionic villus sampling could provide. If the pregnancy continues normally by ultrasound criteria into the second trimester, a judgment can be made whether this of itself would be sufficiently reassuring (perhaps in the setting of all unbalanced forms being very unbalanced), or whether amniocentesis would in fact be desirable. On several levels, each case will have to be assessed on its merits. The CCR will need to be very carefully characterized cytogenetically in the parent and the fetus to ensure accurate prenatal diagnosis; chromosomal microarray is likely to command an increasing role in this setting. The same balanced state identified at prenatal diagnosis raises the same questions, but more pointedly, as in the simple reciprocal translocation (p. 109). By way of example is the CCR 46,XX,t(5;16;10;18) (q13;q22;q11.2;q21) identified at routine prenatal diagnosis in a woman having a history of recurrent miscarriage, reported in Lee et al. (2002), with the same karyotype then being shown in herself. Normal ultrasonography was encouraging, and the pregnancy was continued; at age 2 years, the child was
220
•
Given the very high fraction of embryos expected to be chromosomally unbalanced, preimplantation genetic diagnosis (PGD) would have an obvious attraction, in order to select in favor of the few embryos, if such there be, that might be normal or balanced. We have seen a couple, the husband having a double two-way CCR of karyotype 46,XY,t(2;20) (p25.1;p11.23),t(4;8)(q27;p21.1), who had presented following four first-trimester miscarriages, although their first pregnancy having produced a normal (but unkaryotyped) son. Of 320 theoretically possible karyotypes, only four (1¼%) would be balanced (and thus raising a glimmer of hope that their first fortunate pregnancy might reflect a tendency toward a balanced combination). It was a challenge for the laboratory to develop a sequential FISH strategy, to cover all the possibilities. In the event, following ovulation stimulation with the collection of 25 eggs, of which 23 were subjected to ICSI and 18 embryos resulting, biopsy was achieved in 15 embryos; but none had a balanced constitution. A happier outcome attended the efforts of Escudero et al. (2008) and Lim et al. (2008a), these two groups providing PGD to some eight couples carrying a range of types of CCR: three-way, double two-way, reciprocal plus insertion, reciprocal plus Robertsonian, and others. Four of the couples eventually had a take-home baby. The babies were outnumbered by the created embryos 50 to 1, attesting to the very high genetic risk conveyed by the CCR.
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
13 parental sex chromosome aneuploidy
THERE ARE FOUR MAJOR sex chromosome abnormalities. Otherwise unassisted, infertility is practically inevitable in XXY Klinefelter syndrome and 45,X Turner syndrome, and almost always in Turner variants. The other two conditions, XXX and XYY, apparently have little effect on fertility; furthermore, they are not discernibly associated with any increased risk for chromosomally abnormal offspring. Mosaic forms need to be considered on their own merits.
BIOLOGY We need briefly to consider why X chromosome aneuploidy is associated with so little phenotypic abnormality, compared with autosomal imbalance. The important factor is dosage compensation. Only one X in each cell needs to be fully active. Thus, potentially detrimental effects of an X chromosomal imbalance are mitigated (although not exactly canceled out) by inactivating a supernumerary or abnormal X, or by not inactivating a sole remaining X, as the case may be.
The conceptus with an X chromosome complement in excess of the normal 46,XX or 46,XY accommodates to this imbalance by inactivating any additional X chromosome; or, as Migeon (2007) emphasizes, by maintaining, in each cell, just one X in the active state. This is nearly successful in the 47,XXX female and the 47,XXY male, in whom there is apparently normal in utero survival and a relatively mild postnatal phenotype (p. 478). The fact that some loci are not subject to inactivation, and may therefore function in the disomic (XXY), trisomic (XXX), or even quintasomic (49,XXXXX) states, is likely the predominant reason for the phenotypic abnormalities associated with these karyotypes. In females with abnormal X chromosomes, the pattern of X-inactivation is usually nonrandom, particularly when the imbalance due to the abnormality is “large.” In the 46,X,abn(X) karyotype, with one normal X and one abnormal X—an “abn(X),” as we write it here—the abnormal X is characteristically the inactive one. However, if the abnormality is a •
221
very small deletion or duplication, the inactivation pattern can be random. In the case of the X-autosome translocation heterozygote, the normal X is usually, although not invariably, inactive (Chapter 6). Laboratory Test for X-Inactivation Analyzing the pattern of X chromosome methylation with molecular methodology shows whether inactivation is random or nonrandom. A useful assay is methylation-specific polymerase chain reaction (PCR) based on the androgen receptor gene, located at Xq13 (or any other gene with a convenient polymorphism). A highly skewed pattern, with one X mostly methylated and the other mostly not, is indicative of nonrandom inactivation (Kubota et al., 1999). While this test is performed routinely on a blood sample, there are grounds for believing that the assay result may fairly represent the state in other
body tissues (Bittel et al., 2008). The former tests of Barr body (Fig. 13–1) and late-labeling BrdU analysis (Fig. 13–2) are of historic interest; both assays provide a nice visual illustration of the concept of X-inactivation. DE TA I L S O F M EI OT I C B EH AV I O R
Meiosis proceeds differently in each of the various sex chromosome abnormalities, and each warrants separate consideration. XXX
On theoretical grounds, one might expect the three X chromosomes to display 2:1 segregation, with the production of equal numbers of X and XX ova. But this is not the case. No discernible increased risk for
(a)
(b)
(c)
(d)
FIGURE 13–1 Buccal mucosal cells from (a) a 45,X female, with no Barr body present; (b) a 46,XX female showing the inactive X as a Barr body; (c) a 47,XXX female showing two Barr bodies; and (d) a 48,XXXX female with three Barr bodies. 222
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
(a)
(b)
(c)
(d)
FIGURE 13–2 Partial metaphases showing X-inactivation: (a) a normal X chromosome, (b) an isochromosome of X long arm, (c) an X with a short arm deletion, and (d) a ring X. BrdU had been added for the last 6 hours of culturing. The inactive chromosomes, replicating at this late time in the cell cycle, incorporate BrdU extensively, and thus are palely stained. The active X stains darkly.
chromosomally abnormal offspring of these women has been demonstrated: in the extensive review of Otter et al. (2010), only one case had ever been reported of an XXX mother having had an XXX daughter. Apparently, only normal ova, with a single X, are regularly produced. It may be that the extra X is lost before meiosis occurs (Neri, 1984), with meiosis then proceeding as in the normal XX female. A few instances of premature ovarian failure (POF) in 47,XXX women are on record, including even in adolescence (Holland, 2001). But since XXX and POF are both fairly common, cause and effect remain uncertain. X X Y A N D XXY MOSA I C STAT ES
Barring medical intervention, infertility is almost inevitable in Klinefelter syndrome, although some
remarkable exceptions exist. Terzoli et al. (1992), for example, report an XXY man who had fathered a daughter, with paternity testing confirming fatherhood, and they quote two other such cases. Undetected XY/XXY mosaicism could account for some of these cases. Bergère et al. (2002) showed both XY and XXY cell populations in testicular biopsies from three of four men who, on blood karyotyping and fluorescence in situ hybridization (FISH) analysis, were nonmosaic 47,XXY. These three men had small numbers of sperm identified in the biopsied tissue (one went on to have a child by in vitro fertilization, IVF). Several workers have karyotyped sperm from XXY men, and all find an excess, albeit not a large one, of 24,XX and 24,XY sperm. Possibly, these XY and XX sperm come from XXY spermatogonial stem cells. Alternatively, the abnormal gonadal environment may of itself Parental Sex Chromosome Aneuploidy •
223
predispose to gonosomal nondisjunction in the XY tissue, and from that stance autosomal segregation may also be vulnerable; consistent with this observation is a higher rate of disomy 21 on sperm studies (6.2% vs. 0.4% in controls) (Hennebicq et al., 2001; Bergère et al., 2002). Medical intervention may allow men with Klinefelter syndrome to become fathers. This requires the procedure of testicular sperm extraction (TESE): the surgical opening of the testis, with microdissection of seminiferous tubules under the operating microscope, and analysis on site by an embryologist for the presence of sperm; the procedure is preferably undertaken on the day before programmed oocyte retrieval from the female partner. The few single sperm obtained are injected into the egg (intracytoplasmic sperm injection, ICSI).1 The success rate is variable, with sperm retrieved in about 40%–70% of men, and about half of couples achieving pregnancy (Denschlag et al., 2004; Schiff et al., 2005). The chromosomal outcome for the child appears promising, with only one instance known of fetal XXY (and none of fetal XXX). In one triplet pregnancy, the three karyotypes at chorionic villus sampling were 46,XX, 46,XY, and 47,XXY; fetal reduction was done at 14 weeks, leaving XX and XY twins (Ron-El et al., 2000). However, on examination at a much earlier stage, in the embryo at preimplantation genetic diagnosis (PGD), the rate of aneuploidy, both for the sex chromosomes and autosomes 18 and 21 in particular, may be higher from XXY patients; and this is consonant with the observed higher rate of disomy 21 on sperm study mentioned earlier. Thus, PGD could be offered to these couples (Staessen et al., 2003). As for mosaic states, Giltay et al. (2000) have studied men presenting with severe oligospermia, among whom there were cases of XY/XXY and XY/ XXY/XXXY. Applying FISH to sperm analysis, and probing for chromosomes 18, X, and Y, the aneuploidy rate was somewhat increased compared to a normal population, although in fact similar to a group of normal 46,XY men with oligoasthenoteratospermia who were ICSI candidates. It may be that the sperm abnormalities reflect the testicular defect
per se, rather than being a direct consequence of the XXY constitution. Studies in the XXY mouse support this interpretation (Mroz et al., 1999). XYY
The clinical observation is that XYY men have no discernible increase in risk to have children with a sex chromosome aneuploidy (and XYY or XXY would have been the theoretical risks, from an XYY trivalent at meiosis). A true increased risk of a fraction of a percent could be distinguished only with the greatest difficulty when the background population risk is of a similar order of magnitude. On laboratory study, XYY spermatocytes proceeding through meiosis encounter checkpoints that lead to elimination of most of the abnormal forms (Milazzo et al., 2006),2 but nevertheless, some men may have a small increased fraction of 24,YY and 24,XY spermatozoa in the ejaculate, and in some also, of autosomal disomies. A distinction may be drawn between XYY men presenting with infertility, and those whose fertility is intact, with the sperm aneuploidy rate somewhat higher in the former. Rodrigo et al. (2010) studied the next stage of development, namely, the preimplantation embryo, from five infertile XYY men having had IVF. The rates of chromosome abnormality were double that of a control group, with particular elevations in XY aneuploidy and triploidy. 45,X T UR N ER SYN D R O M E
The great majority of women with 45,X Turner syndrome (TS) are infertile and do not spontaneously menstruate or develop secondary sexual characteristics. The ovaries initially appear to be normal but begin to degenerate in midfetal life. Oöcytes undergo apoptosis and disappear at an accelerated rate and, in most cases, are gone by the age of 2 years: “the menopause occurs before the menarche” (Federman, 1987; Modi et al., 2003). Spontaneous menstruation is uncommon but recorded (Lippe, 1991; Hovatta, 1999), and in one series of eighteen 45,X girls, none had ovarian follicles on biopsy in
1 It is an intriguing thought that, in those cases proceeding to fatherhood through intervention with assisted reproductive technology, the situation may be presented of the homogametic sex being the one to provide the greater quantum of gametes, albeit by a small margin: an extraordinary contrast from the typical vast imbalance due to the heterogametic male of the species. 2 One theory has it that escape from normal meiotic inactivation of Y-borne genes, due to a Y bivalent having formed and thus not able to accompany the X into the sex vesicle, could be the cause of spermatogenic arrest (Turner, 2007).
224
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
childhood or teenage (Borgström et al., 2009). Completed pregnancy in women with an apparent 45,X karyotype is very rare: in a Danish study based on a national TS register, none of 200 45,X women achieved a natural pregnancy (one had twins by ovum donation) (Birkebaek et al., 2002). Sybert (2004) records a total of 18 cases, these women having had 42 pregnancies, of which 17 proceeded to live birth, including one with trisomy 21, and two with 45,X. What is the explanation for fertility in these cases? An obvious point to consider is gonadal mosaicism, with a 46,XX cell line in the ovary. This has often been suggested, but rarely proven (Birkebaek et al., 2002). Jacobs et al. (1997) undertook a systematic search in 84 subjects with TS whose standard blood karyotype was 45,X, with molecular testing of blood and of a second tissue (buccal cells) and found only two cases of X/XX mosaicism. One very thorough study is that reported in Magee et al. (1998b), concerning a 45,X woman who had had seven pregnancies, five miscarrying, one producing a healthy male, and the last terminated following demonstration of fetal cystic hygroma and a 45,X karyotype on amniocentesis. Biopsies of skin, uterus, and ovary at subsequent gynecological surgery all gave a 45,X karyotype, but molecular testing showed two alleles in ovarian DNA, indicating the presence of occult 46,XX tissue. A subtler consideration is whether a pure 45,X oöcyte could in fact proceed through meiosis I, given that the sex chromosome has no homolog with which to pair. X /X X , X/ XX/ XXX, AN D X /X X X MOSA IC ISM TUR N ER SY N DROM E
The relative fractions of the various karyotypes are listed in Table 13–1. For practical purposes, one should make a distinction between those mosaic women who display, to some extent, a TS phenotype, and in whom the fraction of 45,X cells is substantial, versus those of normal phenotype, who have only a low proportion (the latter noted separately later). The risk for chromosomally abnormal offspring hypothetically depends upon the degree and, crucially, distribution of the 45,X cell line. If the gonad contains 45,X cells—in other words, if there is somatic-gonadal mosaicism—and perhaps if these abnormal cells’ survival is enabled by support from surrounding 46,XX oögonia, a true increased risk may exist, although there is difficulty in assessing to
Table 13–1. Relative Frequencies of Turner Syndrome Karyotypes STANDARD MONOSOMY
X mosaicism Isochromosome Xq Ring Deletion Xp
45,X
46%
X/XX, X/XXX, X/XX/XXX 45,X/46,X,i(Xq), 46,X,i(Xq) 45,X/46,X,r(X) 45,X/46,X,del(Xp), 46,X,del)Xp)
7%
Structural abnormality of Y Other
18% 16% 5% 6% 2%
Source: Jacobs et al. (1997).
what extent the ascertainment in published reports has been biased. Sybert (2005) lists 92 pregnancies to 34 X/XX TS mothers, with only 39 of these proceeding to live birth, and six of these babies with an X chromosome abnormality, and one with trisomy 21. In the Danish survey of Birkebaek et al. (2002), 27 out of 78 women with X/XX or X/XX/XXX mosaic states had had at least one child, of whom one was 45,X/46,XY with ambiguous genitalia. Uehara et al. (1999c) record the exceptional circumstance of a woman with 45,X/46,XX having had three monosomic X pregnancies, all showing fetal hydrops; she also had a normal son. The variability of phenotype according to the degree of mosaicism is well illustrated in the report of Lespinasse et al. (1998), who studied monozygous (but not identical) triplets with 45,X/46,XX mosaicism. One child with typical TS had only 6% 45,X cells on blood karyotyping but 99% of fibroblast analysis. One sister with only mild features to suggest TS had 43% of fibroblasts with 45,X, and the third sister, of normal phenotype, had just 3%. Presumably, the mosaicism existed from a very early stage, and the three-way division of the 45,X/46,XX blastocyst, or (if marginally later) of the inner cell mass, happened to cut across an asymmetric disposition of normal/monosomic cells. Low-Level 45,X/46,XX Mosaicism in Phenotypically Normal Women. This category is to be distinguished from that of TS due to 45,X/ 46,XX mosaicism discussed earlier, and it is likely to Parental Sex Chromosome Aneuploidy •
225
be without reproductive consequence. Loss of one X (or one Y) to give an occasional 45,X cell is a normal characteristic of ageing in the 46,XX female (or 46,XY male). Nowinski et al. (1990) studied women who had presented for chromosome testing for a variety of reasons and found X chromosome loss (XCL) to be a consistent age-related phenomenon. Russell et al. (2007) reviewed data from a large number of females having had a peripheral blood cytogenetic analysis and correlated the degree of XCL with age, again documenting a clear association. Up to age 30, 1% or less of cells showed XCL, but rising to an average 2%, 3%, and 5%, at median ages of 40, 50, and 65 years, respectively. To give a sense of where a threshold might lie between normality and the possibility of a significant effect, we may consider the 99th centiles of the fractions of observed XCL, for different cell counts. On a 30-cell count, for example, the 99th centiles were as follows: 6% at 30 years, 9% at 40, 13% at 50, and 17% at 60. These workers also identified an absence of any XCL effect in relation to reproductive loss or infertility, in agreement with Nowinski et al. (although some other studies have contradicted this). Testing a different tissue, such as buccal mucosal cells by FISH, might seem attractive as a means to detect true constitutional X/XX mosaicism, although this may only be useful at higher levels of mosaicism (Schad et al., 1996). X , abn(X) TURN ER SYND R O ME VARIAN TS
Fertility may be retained in some TS variants resulting from an effective partial X monosomy. Deletion of Xp, deletion of Xq, isochromosome Xq, and large ring X are the major categories (Fig. 13–3). In these cases, mosaicism with 45,X or 46,XX cell lines is common. Presumably a partial synapsis occurs at meiosis in the 46,X,abn(X) oöcytes, with the intact segment of the abnormal X pairing with the homologous region of the normal X. A 1:1 segregation would be expected, with equal frequencies of gametes carrying either the normal X or the abnormal X, from that part of the gonad tissue containing the abn(X). X Deletions, pter or qter. Simpson and Rajkovic (1999) summarized the recorded data at that time, with respect to terminal X chromosome deletions, relating the functional ovarian phenotypes, and their summary diagram is reproduced in Figure 13–4. 226
•
(a)
XX
(b)
X
(c)
XXY
(d)
X,r(X)
(e)
X,del(X) (p11)
XY
XXX
XYY
X,i(Xq)
X,del(X) (q13q24)
FIGURE 13–3 Some sex chromosome complements: (a) normal female XX and normal male XY; (b) X and XXX females; (c) XXY and XYY males; (d and e) abnormal chromosomes from females with a ring X, an isochromosome of X long arm, an X short arm deletion, and an X long arm deletion.
Lachlan et al. (2006) reviewed their own experience and the published literature, and they noted fertility with respect to these terminal deletions of the X: p11.4, p21, p21.1, p22.1, p22.12, and p22; and these interstitial deletions: p21.1p11.3 and p22.33p22.12. Transmission from a carrier mother is recorded, and the implications differ according to the gender of the child. Mother-daughter transmission of a del(Xp), and of a del(Xq), are on record and indeed presumed four-generational matrilineal transmission
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
FIGURE 13–4 Terminal Xp and Xq deletions and the associated ovarian functional phenotype, according to position of the breakpoint. (From J. L. Simpson and A. Rajkovic, 1999, Ovarian differentiation and gonadal failure, American Journal of Medical Genetics 89:186–200. Courtesy J. L. Simpson; reproduced with the permission of Wiley-Liss.)
(Wandstrat et al., 2000; Karaman et al., 2003). Palka et al. (1994) describe an apparently nonmosaic 45,X woman who had an abnormal child with an interstitial Xp deletion, del(X)(p22.2::p11.3). Upon restudy, the mother herself had one 46,X,del(X) out of 450 cells, allowing the presumption of a somaticgonadal mosaicism. In a more direct demonstration of gonadal mosaicism, Varela et al. (1991) studied a woman with TS and normal menstruation and who had a 46,X,del(X)(p21) daughter. They showed 5/100 cells with 46,X,del(X)(p21) in one ovary, while all cells from the other ovary, fibroblasts, and lymphocytes were 45,X. Gonadal function can vary in a family, as Zinn et al. (1997) show for a familial del(X)(p21.2). The 45,X/46,X,del(X) mother had three pregnancies, including one miscarriage, and had normal menses till age 39. Her two daughters were both 46,X,del(X). The elder was amenorrheic at age 15, while the younger had spontaneous menarche at age 14½, with regular cycles 1 year later. A very similar family is on record in Adachi et al. (2000). An X deletion may be sufficiently unremarkable in its phenotypic effect in the female
that it is not suspected in the mother, but only discovered fortuitously, such as at prenatal diagnosis (Wandstrat et al., (2000). It is to be noted that some X chromosomes with deletions may actually be combined del(Xp)/dup(Xq), or vice versa (Giglio et al., 2000). Transmission to a male conception will almost always lead to nonviability, due to nullisomy for the segment concerned. In the case of terminal Xp, the limit of viability for the hemizygous deletion male is defined by the cases studied in Melichar et al. (2007), with the maximum survivable loss being ~10 Mb. The phenotype is very abnormal and includes severe psychomotor retardation, Léri-Weill syndrome, chondrodysplasia punctata, ichthyosis, Kallmann syndrome, and ocular albinism. Ring X Chromosome. Rare reports of fertility exist. Blumenthal and Allanson (1997) record a woman with mosaic ring X Turner syndrome, 45,X/ 46,X,r(X), who had been amenorrheic until being given hormone replacement therapy. She had three pregnancies: a healthy 46,XY son, a 12-week Parental Sex Chromosome Aneuploidy •
227
miscarriage, and a healthy daughter with the same 45,X/ 46,r(X) karyotype. The latter was presumably 46,X, r(X) at conception, with postzygotic loss of the r(X) in some tissue. Other such cases are known (Uehara et al., 1997). A rather different example is that in Matsuo et al. (2000), in which a mother and daughter were 45,X/46,X,r(X)(p22.3q28), the ratios of X:Xr(X) being 97:3 in the mother and 73:27 in the daughter. The ring comprised an almost complete X, but small distal Xp and Xq segments were deleted. The two X chromosomes were randomly inactivated, and in consequence, presumably, some “brain genes” would have been functionally nullisomic in those cells having the normal X-inactivated. Thus, mental function in the mother, and more so in the daughter, was compromised. A male with r(X) is almost unknown, but Ellison et al. (2002) describe transmission from a nonmosaic 46,X,r(X) mother to her nonmosaic 46,Y,r(X) son, mother and son both short statured. The breakpoints were very distal, within and beyond the Xp and Xq pseudoautosomal regions, respectively. The “tiny ring X syndrome” due to absence of XIST with a functional X disomy is a quite different clinical entity, typically associated with severe mental retardation, and is discussed on p. 484. Some tiny XIST-lacking ring X chromosomes can yet be associated with only a Turner phenotype, likely reflecting different characteristics, or tissue distributions, of the abnormal chromosome (Turner et al., 2000). 45, X / 4 6 ,XY A N D 4 5,X /47,X Y Y MOSA IC ISM IN TH E M A LE
X/XY mosaicism is occasionally found in males presenting with hypogonadism and infertility with oligo/azoöspermia; in some, the Y chromosome has a deletion at Yq11 (Telvi et al., 1999; Cui et al., 2007). The maleness presumably reflects the fact that the gonad contained XY cells with a functioning SRY gene, that were able to induce effective testicular differentiation with consequent androgenizing capacity. Reddy and Sulcova (1998a) did testicular biopsy on an X/XY man and demonstrated absence of spermatogenesis; about half of the Sertoli supporting cells showed a Y-signal on FISH. One X[10]/XY[90] man with moderate oligoasthenoteratozoöspermia showed a two- to three-fold rate for XY disomy and 18 disomy in sperm (using 18 as a representative autosome) (Newberg et al., 1998). In contrast, a man with 45,X/47,XYY mosaicism reported in Dale et al. (2002) showed normal 228
•
gonosomal complements in 99.9% of sperm. He had presented with infertility due to oligospermia; a normal 46,XY pregnancy was achieved with ICSI. X M I CR O D EL ET I O N S
We confine our discussion to deletions for the demonstration of which molecular cytogenetic and array comparative genome hybridization (CGH) methodology is applied. The risk to transmit the abnormal chromosome will presumably reflect equal segregation, 1:1. If passed from a 46,X,del(X) mother to a daughter, the daughter’s phenotype may be the same as that of the mother (which may well be quite normal). But a firm statement cannot be made. For example, Grillo et al. (2010) identified a novel 1.1 Mb deletion at Xq22.1 in the severely retarded daughter of a mildly retarded mother; X-inactivation skewing, and an influence of the paternal genotype, may have been the basis of the differences in phenotype. If passed from a 46,X,del(X) mother to a 46,Y,del(X) male conceptus, the hemizygous male fetuses will be nullisomic for loci within the region of the deletion. Viability may be possible, but the absence of loci will lead to a “contiguous gene syndrome.” A classic example is the variable combination of Duchenne muscular dystrophy, retinitis pigmentosa, adrenal hypoplasia, glycerol kinase deficiency, and mental retardation, due to microdeletion within Xp21 (Worley et al., 1995). Numerous “neurocognitive loci” are located in proximal Xp, and Qiao et al. (2008) implicate loss of such loci as the cause of autism and intellectual disability in two brothers, due to an Xp11.22 deletion inherited from their mother; a craniofacial phenotype may also accompany this deletion. The carrier female may also display, at least to some extent, the components of a contiguous gene deletion. We have seen a young woman with chronic granulomatous disease, retinitis pigmentosa, and ornithine transcarbamylase (OTC) deficiency, having an Xp deletion shown on FISH, these loci being within about 1 Mb at Xp11.4p21.1. She suffered recurrent upper respiratory tract infections and had a history of surgery for mastoid osteomyelitis and lung abscess; her peripheral vision was poor; and she unconsciously self-managed the OTC deficiency by avoiding high-protein foods. X-inactivation was random, on blood analysis. She came to prenatal diagnosis and elected to terminate a male pregnancy with the deletion, the predicted phenotype being severe (Coman et al., 2010b).
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
X DU P LIC ATIO N S
In the rare case of the abnormal X having a duplication of X material, 1:1 segregation in the female heterozygote would be expected. Offspring inheriting the abn(X) are hemizygous males or heterozygous females. Duplications may be “classical,” and detectable cytogenetically, or “microduplications,” requiring molecular methodology for their recognition. The basis of the rearrangement can be a direct duplication, an inverted duplication, or an isodicentric chromosome ( James et al., 1997; Shapira et al., 1997a; Matsuo et al., 1999; Kokalj Vokac et al., 2002). The typical karyotype is 46,X,dup(X), although mosaic forms are on record, such as 46,XX/46,X,dup(X) and 45,X/46,X,iso dic(Xq). Hemizygous sons, in whom the abn(X) is genetically active, have a functional partial X disomy, and are of abnormal phenotype, often severely so. Two notable long arm duplications are Xq21.1–q21.31 and Xq27.2–qter, both leading to a clinical phenotype resembling Prader-Willi syndrome, and which can be transmitted from a carrier mother, who may or may not be phenotypically normal herself (Sanlaville et al., 2009). Gabbett et al. (2008) describe a boy with poor motor and language development, who became a “food seeker” as an infant, and in whom a dup(X)(q21.1–q21.31) was identified. His mother, who had had “learning difficulties” at school, proved (on blood analysis) to be a mosaic carrier of the duplication, with a random pattern of X-inactivation. The phenotype in heterozygous daughters is less predictable. If the rule of selective Lyonization holds, the abn(X) is consistently the inactivated one, and normality might, in theory, be expected; while if the rule fails, random inactivation could, in theory, lead to an attenuated functional partial disomy, with phenotypic abnormality. Thus, Apacik et al. (1996) report a normal grandmother and a mother with a duplication of Xq12-q13.3, due to an inverted insertion. In them, the abn(X) was preferentially inactivated. The family only came to attention when the mother had two retarded and dysmorphic sons with 46,Y,dup(X). Tzschach et al. (2008) report a similar circumstance in families segregating a dup(X)(p11.3p21.1) and a dup(X) (p22.11p22.2): the males severely retarded, the females all healthy. Numerous examples exist of random X-inactivation associated with abnormality (Matsuo et al., 1999; Monnot et al., 2008). Armstrong et al. (2003) describe an abnormal child
with 46,X,dup(X)(q22.3q26) in whom the dup(X) was preferentially inactivated, but with a part of the duplicated segment apparently escaping inactivation. They propose functional disomy restricted to this small part to have been the cause of the observed anomalies. Somewhat similarly, Kokalj Vokac et al. (2002) showed consistent inactivation of the dup(X) in a girl with a de novo 46,X,dup(X) (p11.23-p22.33::p11.23-p22.33), except for the intriguing observation of early-replication within the actual breakpoint region; this may have been the basis of her abnormal phenotype. But theory quite often does not apply in practice, and instances are recorded of random X-inactivation with associated normality, and preferential inactivation in the setting of phenotypic abnormality (Matsuo et al., 1999). Stankiewicz et al. (2005) report a dup(X) (q26.2q27.1), a 7.5 Mb segment, passed from mother to daughter, and both phenotypically abnormal, in spite of skewed inactivation. Interstitial microduplication of the region on Xq28, which includes the MECP2 gene (Lubs syndrome), is noted on p. 332. A recurrent microduplication, which includes, but is not necessarily confined to, the segment Xp11.22-p11.23, has the curious quality that in most affected females, it is the normal X that is preferentially inactivated (Holden et al., 2010). Mental retardation, borderline to severe in nature, may affect males and females similarly. The phenotype is notable for including a particular abnormal electroencephalographic (EEG) pattern, in the absence of obvious seizures. The inheritance could be described, in most, as X-linked dominant; for the smallest duplication, transmission follows an X-recessive pattern. SE X CH R O M OS O M E PO LYS O MY
The 48,XXXX female characteristically has diminished ovarian function, and fertility in pure XXXX is on record in only one case (ascertained through a Down syndrome child) (Gardner et al., 1973b). Sterility is presumably invariable in XXXY and XXYY males, who have a further sex chromosome superadded upon the Klinefelter karyotype (Linden et al., 1995). Y C H R O M OS O M E A B N O R M A L I TY
Y chromosome deletions associated with male infertility are discussed in Chapter 23. A single family is Parental Sex Chromosome Aneuploidy •
229
recorded with an interarm insertional Yq duplication, presumed transmitted from a normal father to two normal sons. The wife of one had presented with two miscarriages, which may or may not have been related (Engelen et al., 2003).
GENETIC COUNSELING Many of the gonosomal disorders are associated with infertility, or at least subfertility. Some present a phenotype of relatively mild abnormality. Whether prenatal diagnosis is chosen, in those who are able to achieve pregnancy, may depend on the parents’ perception of the seriousness of the potential abnormal outcome. Their decision may well also be influenced by how difficult it was to achieve the pregnancy. Inference from prenatal X-inactivation analysis, in abn(X) cases, may be fraught with uncertainty. XXX
XXX mothers have no discernibly increased risk of bearing chromosomally abnormal children. A theoretical increased risk for children with an X aneuploidy has not been demonstrated in practice. Despite reports of chromosomally abnormal children born to XXX women, it should be emphasized, as did Dewhurst and Neri in 1978 and 1984, respectively, that when biased ascertainment is taken into account, no excess of abnormal offspring has been reported. Near-silence subsequently in the literature on this issue suggests at least a rarity of abnormal pregnancy outcomes; one such case, an XXX daughter of an XXX mother, is mentioned in passing in Haverty et al. (2004). An additional risk estimate of <½% for a chromosomally abnormal child may be reasonable. A possibility of premature ovarian failure with 47,XXX can be brought to the attention of these women, which may assist in decisions about the timing of childbearing. XXY
Hardly ever will these men father children, without recourse to IVF (see earlier). The early data are certainly small (Fullerton et al., 2010), but one may propose an approximate risk figure of 2% for a sex chromosomal abnormality in the child. Sperm and PGD chromosome studies indicate that as well as this small increased risk of gonosomal aneuploidy, autosomal aneuploidy might also be implicated, 230
•
albeit that an actual case in a child is yet to be observed (Staessen et al., 2003). In younger men in whom the diagnosis of XXY is made, and who do have sperm in the ejaculate, gamete banking may be appropriate (Schiff et al., 2005; Ichioka et al., 2006). Indeed, given a greater success rate in younger men, Ferhi et al. (2009) advise that the procedure is best undertaken before the age of 32 years, which they see as a critical cutoff. A concomitant AZF deletion (p. 391) may warrant checking, and particularly in mosaic men with azoöspermia (Mitra et al., 2006). XYY
To our knowledge, there is no report of a discernibly increased risk for the XYY male to have chromosomally abnormal children. A slight increase in gonosomal imbalances in sperm (see earlier) might nevertheless lead some to choose prenatal diagnosis. The risk might be greater in those XYY men who need fertility treatment (Wong et al., 2008), and for whom PGD might therefore be appropriate. 45,X T UR N ER SYN D R O M E
Natural fertility is very rare. However, a 45,X woman who has spontaneous menses may possibly be fertile. Endocrine and ultrasound studies may clarify whether ovulation is occurring, or likely to occur (Mazzanti et al., 1997; Paoloni-Giacobino et al., 2000a). Any period of fertility is likely to be shortlived; thus, a woman with 45,X TS who wishes to have a child should not delay in trying for a pregnancy. Tarani et al. (1998) reviewed the literature on pregnancy outcome in (apparently) nonmosaic TS women. In all, fifteen 45,X women had 26 recorded pregnancies, with 9 miscarriages and 16 completed pregnancies. From these 16 pregnancies there were 13 normal children (81%), 2 stillborn, and 1 with Down syndrome. In further data from Sybert (2005), pregnancies with 45,X and 46,X,del(X) are listed. While the miscarriage rate is high, it may be that the maternal gynecology was more contributory than were fetal factors. There may have been selective reporting in the literature of those with abnormality (although it is true that any natural pregnancy in a 45,X woman might warrant publication), but nevertheless the conclusion of an increased risk for a chromosomal abnormality seems inescapable.
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
For the great majority of TS patients who cannot make their own eggs, ovum donation with IVF may be one route to achieve childbearing (Hovatta, 1999). Foudila et al. (1999) report their experience with 18 women with TS, and although the rates of embryo transfer were similar to those of other women with primary ovarian failure, the miscarriage rate was high (40%); possibly, this may have been due to uterine factors. Bodri et al. (2009) report a similarly discouraging experience. Fénichel and Letur (2008) insist on the advisability of transferring a single embryo only. Any genetic risk to the TS patient bearing children via ovum donation is due to that of the biological donor parents. A related donor (mother, sister) would have obvious attraction, and the improving methodology of ovum storage offers the possibility of maternal donation well ahead of the time of potential use (Schoolcraft et al., 2009). Gidoni et al. (2008) report a 33-year-old mother having “oöcyte vitrification” for the potential use of her daughter, with isoXq Turner syndrome. These authors discuss the ethical issues involved and conclude that the procedure is reasonable and acceptable, with the mother’s motives purely altruistic, and she “is simply providing an option for her daughter.” Anticipating possible artificial fertility, the offer should be made of hormone treatment from the age of 10–12 years, in order to avoid uterine hypoplasia (Leclercq et al., 1992). Otherwise, obstetric management must take account of possible cardiovascular complication (Fénichel and Letur, 2008). MOSA IC 4 5 ,X TUR N E R SY N DROM E
Women with 45,X mosaicism and a TS phenotype presumably carry the 45,X cell line in much of the soma and gonad. Categories include X/XX, X/ XXX, and X/XX/XXX. Ovarian function is often intact, although premature failure is common (Blair et al., 2001; Sybert, 2005). The risk for miscarriage is increased (Homer et al., 2010). There is apparently an increased risk for X monosomy in a child, and this is consonant with theoretical expectation (Sybert, 2005). Normal cells in the gonad may provide support for monosomic cells that otherwise would not have survived. The upper limit of the risk may be about 15% (Tarani et al., 1998). In those having suffered ovarian failure, ovum donation may
succeed.3 A few may, as adolescents, have suitable follicles available for biopsy and thus be candidates for attempted ovum vitrification for possible future use (Borgström et al., 2009; Lau et al., 2009). Low-Level 45,X/46,XX Mosaicism in Phenotypically Normal Women This category is, for practical purposes, to be distinguished from that of the preceding section on “Mosaic 45,X Turner Syndrome.” The discovery of a low-level (a singledigit percentage) of 45,X cells in a woman presenting no phenotype traits of TS is not to be overinterpreted, nor is a reproductive risk to be exaggerated. Indeed, no such risk may apply (Horsman et al., 1987). Loss of an X chromosome is a normal concomitant of ageing (see also “Biology” section). X ,ab n (X) T UR N ER SYN D R O M E VA R I A N T
Not infrequently, women with incomplete Turner phenotypes due to a 46,X,abn(X) karyotype have normal secondary sexual development, and fertility is likely or indeed proven. The majority involve deletions of Xp or Xq, and ring X chromosomes. The deletions may be of quite substantial size, with a phenotypic range from partial TS through minor menstrual abnormality. Premature ovarian failure is likely, and it is practical advice that childbearing should be embarked upon earlier rather than later. Assuming 1:1 segregation (a fair assumption), the deleted X will be transmitted in 50% of ova. In Tarani et al.’s (1998) review, examples are given of TS mothers with Xp deletions and ring X chromosomes, most of whose children (5 out of 6) were abnormal, with the same type of TS. It might be guessed that this weighting toward abnormality was due to publication bias. If the ovum containing a deleted X meets an X-bearing sperm, a conceptus with the same karyotype as the mother results. If the ovum meets a Y-bearing sperm, a zygote with partial nullisomy X results, and it may end in abortion, depending on the size of the deletion. Viable offspring are in the ratio of 1:1:<1 of chromosomally normal males, normal females, and X,abn(X) females. Mosaicism in the mother may well be reflected in mosaicism in the daughter. Uehara et al. (1997) record a
3 Ovum donation might be too successful. Makrakis et al. (2009) report a young woman, X/XX, who had three embryos from donated ova transferred, with all three implanting, and one dividing to give a monozygous pair: a quadruplet pregnancy. She underwent fetal reduction, and eventually she gave birth to twins.
Parental Sex Chromosome Aneuploidy •
231
45,X/46,X,r(X) mother to whose child she transmitted the ring chromosome; the ring was present in 4% of the mother’s cells and in 34% of the daughter’s. A similar case is reported in Blumenthal and Allanson (1997), in this instance the mother having been on hormone replacement therapy.
chosen, should be a molecular genetic exercise. Measurement of X-inactivation would not offer a clear interpretation of risk adjustment, in the case of transmission of the abn(X). Preimplantation genetic diagnosis may be appropriate. X D UPL I CAT I O N
X /X Y M A LE
Infertility is probable. If there is any sperm production, an IVF pregnancy might be possible. Given a possible increased risk for aneuploidy, gonosomal or autosomal (Newberg et al., 1998), PGD is to be considered in that setting.
The female carrier has a chance of 50% to transmit the normal X, and a risk of 50% to transmit the dup(X). If an abn(X) pregnancy proceeds to live birth, a son would be abnormal due to X disomy. A daughter is not necessarily protected by selective inactivation, and thus phenotypic abnormality is possible.
X M I CRO D ELETIO N
The carrier can be fertile, and 1:1 segregation with respect to the normal X and the abn(X) is to be expected. This implies a 50% risk to have a son affected with the full “contiguous gene syndrome” or a daughter who might display a partial phenotype. There may be an increased fetal loss rate with the 46,Y,del(X) karyotype. Prenatal diagnosis, if
232
•
SE X CH R O M OS O M E PO LYS O MY
Many XXXX women are of low-normal or borderline intelligence, and the questions of fertility and genetic risk may well be raised by their carers. In fact, it appears that sterility is usual. XXXY and XXYY men are undoubtedly sterile.
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
14 parental autosomal aneuploidy
A QUESTION OF REPRODUCTION is usually an academic matter in individuals with a cytogenetically detectable functional autosomal aneuploidy. But toward the milder end of the phenotypic range, social and emotional development may be such that forming a stable relationship is possible. Some who lack that degree of maturity may yet have a social freedom that opens the possibility of a sexual encounter. Either on their own behalf, or through the agency of parents or other carers (whose agenda may include sterilization), such people may present to the genetic clinic. Ethical issues raised in this context are aired in Chapter 1. With some of the deletions and duplications of lesser genetic imbalance revealed by molecular techniques, the phenotypic effects may be very mild, with social and intellectual functioning well within the normal range. The mosaic state may on occasion be associated with an apparently normal phenotype, or at least very close to normal. Some of these persons will have been identified by their having had a child with the imbalance for which they themselves have a
low-level mosaicism. Some present with infertility. The theoretical risk will depend upon the extent to which the gonad carries the aneuploid line. In contrast, some structural imbalances are without discernible phenotypic effect. Deletion or duplication of a small segment of euchromatin, or the presence of a supernumerary marker chromosome (SMC), is occasionally recognized fortuitously in normal and fertile individuals.
BIOLOGY Parental Trisomy Parental Trisomy 8 Mosaicism. Mosaic trisomy 8 arises postzygotically, from an initially normal conceptus (Robinson et al., 1999). Habecker-Green et al. (1998) review reports of reproductive status in 46/47,+8 individuals, and there is only a tiny number of cases, usually in persons in whom the diagnosis would not have been suspected clinically. They describe a woman with mosaic trisomy 8 •
233
having a history of four spontaneous losses, including a 46,XX fetal death at 27 weeks; her next pregnancy produced an apparently normal 46,XX daughter. Rauen et al. (2003) report a woman who presented a more typical clinical picture of trisomy 8 mosaicism having a 46,XX child (phenotypic abnormality in the child probably reflected paternal characteristics). Mercier and Bresson (1997) studied an otherwise healthy man, whose partner’s recurrent miscarriage was the presenting problem, and in whom the peripheral blood karyotype was 46,XY[92]/47,XY,+8[8]. On fluorescence in situ hybridization (FISH) analysis of 25,000 spermatozoa, 398 (1.6%) showed disomy 8, which compared with a rate in control sperm of 0.2%. It is perhaps surprising that such a low level of disomic 8 sperm should be associated with a high miscarriage rate (always assuming that the link is causal and not coincidental). We have seen a somewhat similar case, a man of above-average intelligence and excellent physical health, with infertility due to oligospermia, in whom low-level trisomy 8 mosaicism was shown on two separate blood samplings; in his case, one could not exclude that the abnormal cell line was confined to hematological tissue, and the oligospermia coincidental. Parental Trisomy 18 Mosaicism. This is extremely rarely recorded in adulthood, and Tucker et al. (2007) review in detail the range of phenotypes. Some had presented with a history of miscarriage, and some due to having had a child with trisomy 18. Because of the usual high rate of lethality of trisomy 18 in utero, the genetic risks obtaining in such persons would apply substantially to miscarriage. The risk will relate to the gonadal load of trisomic cells; this is not usually known, but some gametic studies are recorded. Bettio et al. (2003) report a woman of normal intelligence with 70% trisomic 18 cells on blood but none on fibroblast karyotyping, presenting with infertility. Ovarian biopsy showed 90% trisomic cells from right ovarian biopsies and a normal karyotype in left ovarian tissue. A man of normal intelligence and appearance, presenting with severe oligospermia, had approximately 50% trisomy 18 mosaicism on blood and buccal mucosal cell analysis, although only 3% in skin fibroblasts: on sperm study, there was a 10-fold increase in disomy 18, compared with control data, although the absolute fraction was small (0.68%) (Perrin et al., 2009). Both testes may be free of the trisomic line, as apparently in the father of a normal 234
•
daughter described in Lim and Su (1998). He was of normal intelligence and worked as a sales representative, and had “slightly unusual facial features.” The trisomic line was found only in blood (76%) and not in skin fibroblasts, and the disomic 18 rate in sperm was similar to that of a control. Maternal Trisomy 21. At female meiosis, the classical scenario is that the three homologs form either a bivalent and a univalent, or a trivalent (Fig. 14–1) (Wallace and Hultén, 1983). If the former, the bivalent may disjoin and segregate symmetrically, but the univalent passes at random to either daughter cell (1:1 +1 segregation). If the latter, a trivalent may of itself set the stage for aberrant segregation (2:1 segregation). In either case, the result is disomic (24,+21) and normal (23,N) gametes in equal proportions. Speed (1984) has observed trivalents in about 40% of meiotic cells and a bivalent plus a univalent in the remaining 60%. An alternative scenario is that the “third” chromosome 21 separates prematurely into chromatids, and each chromatid then passes to a daughter cell (the oöcyte, and the first polar body). Cozzi et al. (1999) provide direct evidence for this mechanism in the FISH study of unfertilized oöcytes from a woman who was presumed to be a 46/47,+21 gonadal mosaic. In a review of the literature, Shobha Rani et al. (1990) list 30 reports of pregnancy in Down syndrome (DS) women. The ratio of DS to normal offspring was 10:17 (there were three abortions), not significantly different from a 1:1 ratio, but suggestive of a deficit in trisomic offspring. A reasonable interpretation is that 46,N and 47,+21 conceptions occur with equal frequency, but loss of pregnancy is greater with the trisomic fetuses. About one-third of the
FIGURE 14–1 Possible synapsis of three no. 21 chromosomes: (a) as a trivalent and (b) as a bivalent and a univalent.
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
46,N offspring were nevertheless abnormal, which may have reflected paternal or environmental factors. Cunniff et al. (1991) noted a diminution in the number of oöcytes in the ovaries of DS girls at the time of birth, which could be the cause subsequently of subfertility. Paternal Trisomy 21. Spermatogenesis is reduced in the male with DS, but it does not necessarily fail; and a tiny number of examples of proven or suspected fatherhood in DS males have been documented (Pradhan et al., 2006). Parental Trisomy 21 Mosaicism. In practice, it is usually only those recognized mosaic individuals with a low percentage of +21 cells who seek genetic advice. These people typically come to notice because they are studied as apparently normal parents of more than one DS child (and see p. 282). The important factor, if it could only be known, is the degree to which the gonad comprises 46,N and 47,+21 cells. The trisomic cells (see earlier discussion) produce disomic and normal gametes in equal proportion; of course, normal cells, other things being equal, give rise only to normal gametes. Thus, the proportion of abnormal gametes produced depends on the proportion of germ cells that are trisomic. In the limit, the gonad might be fully 47,+21. Any level of correlation between the degree of mosaicism in lymphocytes and gametes is not readily amenable to study. Familial trisomy 21 mosaicism is on record but is exceptional (Kovaleva, 2010).
Parental Partial Aneuploidy (46,rea) Uncommonly, the unbalanced and phenotypically abnormal carrier of a classical cytogenetically detectable chromosomal rearrangement may be functionally fertile. The usual forms are deletion, duplication, and the derivative chromosome from an unbalanced translocation. Barber (2005) lists the recorded deletions and duplications that have been transmitted by partially aneuploid parents, with all but two of the autosomes represented at least once, and chromosomes 4, 5, 8, and 18 the most often seen (Table 14–1). Microarray comparative genomic hybridization (CGH) methodology is revealing many more examples of imbalance in which, due to the lesser extents of the loss or gain, a phenotype of lesser severity may more frequently allow the possibility of the heterozygote passing on the abnormality. While the rules of mendelian segregation impose a
straightforward 1:1 ratio of normal:abnormal chromosome transmission, there is the potential complicating factor of variable expressivity, and indeed nonpenetrance, which might in some cases be due to the presence or absence of other imbalances elsewhere in the genome (Veltman and Brunner, 2010), and which we discuss also in Chapter 17. Some microarray-level imbalances are achieving syndromic status, such as del(3)(q29), which is noted in Chapter 19 (p. 312), but which could also have a rightful place, as a parental autosomal imbalance, in this chapter. It is often observed that the child is more severely affected than the parent. In part, this will be due to bias of ascertainment: parenthood is more probable in those whose abnormality yet allows them to have acquired a degree of social functioning, whereas a more markedly affected child will likely come to medical attention, and only then is the parental state recognized. Another consideration may be the “genetic quality” of the other parent, and bearing in mind the strong tendency toward “positive assortative mating” with respect to intelligence. (Of course, any diminished “genetic quality” need not apply, if the other parent were intellectually disabled due to environmental factors, such as birth injury.) CLA SS I CA L D EL ET I O N S
Rarely, the parental phenotype may be severe, such as the cri du chat syndrome documented in mother and child in Martínez et al. (1993; and see p. 18). Loss of a tumor suppressor gene, the APC gene which is the basis of familial adenomatous polyposis, accompanied a del(5)(q22q23.2) in a retarded man, his aunt, and inferentially his retarded mother, causing polyposis in at least the man and his aunt; other family members carried a balanced insertional rearrangement (Cross et al., 1992). A less severe clinical picture is more often observed, as may be exemplified by deletion for the segment 8p23.1-pter in Pettenati et al. (1992); a family photograph shows unremarkable physical appearances, but the del(8p) children had learning and behavioral difficulties. The well-known chromosome 22q11 deletion can be transmitted from parent to child, with a different clinical picture in each (p. 327); typically the child is more severely affected than the parent. Another well-recorded abnormality, the 18p deletion, has been reported due to familial transmission in six families (Maranda et al., 2006); and similarly, a number of instances are known of the parental Parental Autosomal Aneuploidy •
235
Table 14–1. Euchromatic Deletions or Duplications That Are on Record in the Literature as Having Been Transmitted from Parent to Child, and Having an Associated (and Presumably Causally Associated) Abnormal Phenotype REGION
deletion: 1q42.1q42.3 2p11.2p12 3p25pter 4p15.2p16.1 4q33qter
ASCERTAINMENT
Developmental delay, ADD Wilms tumor, developmental delay Speech delay
5p14p15.3 5p13.3p14.3 5p13.1p14.2
MR, dysmorphism Multiple congenital abnormalities and language delay Developmental delay Developmental delay and dysmorphic features Developmental delay and dysmorphic features Developmental motor, speech delay Speech delay, dysmorphic Cat cry at birth, low birth weight Speech delay, hearing loss, mild MR Speech delay, mild developmental delay Speech delay, raspy voice Speech and developmental delay Multiple congenital abnormalities Cri du chat Microcephaly, small Speech delay
5p13p15.1
Maternal age
7q11.23 8p23.1pter 9q31.2q32
Williams syndrome Mental slowness, seizures Developmental and growth delay, unusual appearance Developmental delay Leukocoria Developmental delay MR Previous son with MR MR, short stature
4q33q35.1 4q33q33 4q32q33 5p15.32pter 5p15.31pter 5p15.3pter 5p15.3pter 5p15.3pter 5p15.3pter 5p15.3pter 5p15.1pter
11q24.2qter 13q14.1q21.3 14q31q31 15q11q12 18p11.3pter 18p11.21pter
REGION
ASCERTAINMENT
duplication: 1q23q25 2q24.3
Mild MR and dysmorphism Epilepsy
3q25.3q26.2 4q31.22q33 4q31.1q32.3
Microcephaly, congenital heart disease, and deafness Mild MR and dysmorphism Developmental delay, nasal speech
5q15q22.1 7p12.2p13
Hyperactive, mild MR Failure to thrive
7p12.1p13
Short stature, ?Silver-Russell
7q32q36.1 8p23.1p23.1 8p23.1p23.1
Developmental delay, behavioral problems Congenital heart disease Developmental delay
8p23.1p23.1
Developmental delay, hypotonia
8p22p23.1
Mild MR only
8p21.3p23.1 8p21.3p23.1
Congenital heart disease Speech delay
8p21.3p22 (or p22p23.1) 8p12p21.1 9p22p24 10p13p15
MR, short stature, hypertelorism
11q13.5q21 (or q21q23.1) 14q13q22 15q11.2q13 15q11.2q13 15q11.2q13 15q11q13 16q11.2q12.1 18 cen-pter 22q11.2 21q22qter
Developmental delay Short, low IQ, dysmorphic Developmental delay, especially speech Maternal age Developmental delay Developmental delay, hypogonadism Severe MR Developmental delay Developmental delay Speech delay Dysmorphic, moderate MR Variable (p. 327) Unusual appearance
Table 14–1. continued REGION
deletion: 18p11.2pter 18p11.23pter 18p11.2pter 18p (pre-banding) 18q23qter 18?q21qter 18q22.3qter 20p11.2p12.2 21q11q21.3 22q11.2q11.2
ASCERTAINMENT
Abnormal ultrasound Multiple congenital abnormalities MR, short stature Failure to thrive, ptosis Dysmorphic Multiple congenital abnormalities Phenotypic abnormality Dysmorphic Dislocated hips Cardiac failure
ADD, attention-deficit disorder; MR, mental retardation; ?, suspected diagnosis. Source: From the review of Barber (2005), with the addition of del 7q11.23, dup 2q24.3, and dup 22q11.2.
transmission of the 7q11.23 deletion of Williams syndrome (Metcalfe et al., 2005). As a rather obvious rule, the smaller the deletion, the milder the phenotype: Sanford Hanna et al. (2001) describe a very small distal 1q deletion, del(1)(q42.1q42.3), in a mother and son with rather minor physical anomalies, the mother requiring “assisted living services,” and the son attending special education classes at the age of 13, working at approximately a third-grade level. CL ASSIC AL D UP LIC AT I ON S
Transmitted duplications are slightly less often reported than are deletions. This lower frequency may represent an ascertainment bias based on microscopy because only large duplications that involve more than one band can be easily visualized. Those duplications that result in a slightly broader band are less likely to be seen, as compared to deletions that remove an entire band or a substantial part of a band. Based on array-CGH, duplications less than 500 kb are commonly observed, and they are usually inherited from a carrier parent (see Chapter 17). Three-generation transmission of a dup(7) (p11.2p12) is recorded by Leach et al. (2007) in three affected family members having a “mild cognitive deficiency.” Glass et al. (1998) report a mother and daughter with a dup(2)(q11.2-q21.1), due to an (8;2) insertion, leading to a clinical picture of minor facial dysmorphism, short stature, mild intellectual deficit, and psychiatric disease (schizoaffective disorder in the mother, paranoid psychotic state in the
daughter). With an insertion such as this, the genotype is presumably that of a pure 2q partial trisomy. An example of the parent being less affected than the child is given in Pazooki et al. (2007), the mother having a milder dysmorphism than her mentally retarded daughter, and both 46,XX,dup(6) (q21q22.1). We have seen a family with, inferentially, four-generation transmission of a duplicated chromosome (segment 10p14). One great-grandparent, we may guess, was a gonadal mosaic, and then in two separate branches of the family the duplication was passed down, causing considerable psychological and social morbidity as it did so (Voullaire et al., 2000a). (Only when the cytogeneticist gathered three families with the same dup(10p) for a study was it realized that they were, in fact, related.) Somewhat similar is the family in Arens et al. (2004), a four-generation family, but in this case there were some individuals with a balanced 3;5 translocation, three of whose offspring with an unbalanced form, producing a duplication of 5q22.1–5q31.3, went on to transmit the same imbalance to their children. Genomic imprinting was an important factor influencing the effect of a familial ins(2;6)(p22.2; q22.33q23.3) which had been transmitted through three generations (Temple et al., 1996), discussed also on p. 359. A child with transient neonatal diabetes had a duplication for 6q22-q33 due to the insertion, which she had inherited from her father. He and his mother also had the same unbalanced karyotype, but neither had any history to suggest neonatal Parental Autosomal Aneuploidy •
237
diabetes. It was the imprinting effect due to paternal transmission that resulted in the child’s medical condition. dup(15)(q11.2q13). Proximal 15q is a vexing region, because of the need to distinguish between harmless euchromatic variants (see p. 264) and a pathogenic dup(15)(q11.2q13). The distinction is to be made upon the absence or presence, respectively, of the Prader-Willi/Angelman critical region (PWACR). There is the added complication that this region of chromosome 15 is subject to imprinting, with phenotypic abnormality, including autism spectrum disorder, typically occurring in the setting of transmission from the mother; a milder phenotype extending into the normal range may apply to paternal transmission (Bolton et al., 2001) (see also p. 329). A complex story is that of the family in Gurrieri et al. (1999). A child with pervasive developmental disorder, atypical autism, and epilepsy had, on molecular analysis, a dup(15) (q11.2q12) of ~2 Mb size; his mother had a duplication in the same region, but not so extensive (~1 Mb), and the grandparents had normal chromosomes. Thus, the first duplication had arisen on transmission from the normal grandfather, and then on transmission from the normal mother to the affected child, the duplication expanded. The child’s abnormality is likely due to aberrant expression of imprinted genes. MICROARRAY-DETECTABLE IMBALANCES
Microarray testing (or other molecular methodology) casts a wider net, and we mention only a very few of the rapidly growing list of examples in which parental transmission is known. Yu et al. (2008) identified two families with a duplication of the DiGeorge region, that is, dup(22)(q11.2): in one family of size 3 Mb, and in the other, 1.5 Mb. The phenotypes differed quite widely through each family, and indeed in one, the normal grandmother acted as carer for two of her disabled grandchildren. A little telomeric of this site is the microduplication of 22q11.21q11.23, most cases of which turn out to have been inherited from a parent, and most of these parents are judged to be “apparently normal” (Coppinger et al., 2009b).The Williams syndrome region, 7q11.23, is another which, when present as a 238
•
duplication, is not infrequently observed in parent– child transmission (Van der Aa et al., 2009). A number of other examples are on record of variable phenotype, and of inheritance from normal parents (Wentzel et al., 2008; Ou et al., 2008b). Autism is an area of active research in the microarray laboratory. Microduplication of 16p11.2 is a microarray-level imbalance that has been associated with autism spectrum disorders (Fernandez et al., 2010), although others argue that the associated neurocognitive profile may comprise only nonspecific components that can also be observed in, but not necessarily adding up to, autism (Rosenfeld et al., 2010). This microduplication, and occasionally the countertype microdeletion, can be transmitted from a parent, who may or may not show a cognitive phenotype, and may also be seen in other apparently unaffected family members. The dup(15) (q11.2q13), as mentioned earlier, has an association with autism, and familial autism with a segregating microduplication in this region is described in Van der Zwaag et al. (2010); but the complexity of interpretation of these small genomic lesions is illustrated by the fact that this same microduplication is also seen in a general population, albeit at a lower frequency than in autistic patients. By no means is this the only such example (among many others, see also del(1)(q21.1), p. 311), and interpreting the parental transmission of a genomic imbalance that might or might not be pathogenic is turning out to be a particular challenge as microarray analysis becomes widespread. A familial phenotype restricted to a single trait, or traits restricted to a single system, might suggest the imbalance of a single important gene, and if this is due to deletion or duplication, molecular analysis may be revealing. Thus, Veenma et al. (2010) studied a large family in which a 15q26.3 microdeletion cosegregated with short stature (the methodology in this case being MLPA). The only notable gene deleted was IGFR1, a growth-related gene. The family described in Heron et al. (2010) manifested dominantly inherited neonatal seizures and intellectual disability, and these workers identified a 1.6 Mb duplication at 2q24.3, this region containing a “gene family” of three sodium channel loci. Triplication. The rare case of a transmitted triplication is exemplified in Wang et al. (2004), in their report of a mother and her three sons all with 46,trp (4)(q32.1q32.2), and all displaying neurocognitive compromise and minor dysmorphism. Two of the
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
boys had brain imaging, which showed increased size of the ventricular spaces, denoting a reduction in brain substance.
Mosaicism If a normal cell line coexists with an abnormal karyotype—in other words, there is a 46,N/46,(abn) mosaic state—a high risk for abnormal pregnancy may be implied, an essentially normal parental phenotype notwithstanding. Yip et al. (1996) describe a normal woman with 46,XX/46,XX,der(6)t(6;8) (q27;q22.2), who had presented with recurrent miscarriage. A balanced 6;8 translocation had presumably arisen at a somatic mitosis, and subsequently an unbalanced der(6) cell line was generated. This cell line likely contributed to ovarian formation, and the miscarrying pregnancies were due to conceptions from 23,der(6) ova. If the abnormal cell line in the parent is predominant, his or her phenotype will be abnormal, as exemplified by the mother reported in Cox et al. (2002) having the karyotype 46,XX,dup(7) (p15.3p22)[75]/46,XX[15], and whose abnormal son had the duplication in nonmosaic state. Very rarely, there can be parental mosaicism with two abnormal cell lines having the opposite imbalance, and a child can be born with one of these imbalances in nonmosaic state (de Pater et al., 2003b). PA R ENTA L PA RT I A L A N E U P LO I DY D U E TO SM A LL S UP ERN UM E R AR Y M AR KE R CHR O MOSOME OR EXTR A STR U C TU R AL LY A BNORMA L C H ROMOS O M E ( 4 7 , + s SM C , 47, + ESAC)
The concept and nomenclature of the SMC has evolved along with the increasing precision which, over the decades, the cytogeneticist has been able to apply to chromosome analysis. The description “marker” originally denoted an additional chromosome whose origin could not be determined. With banding, larger markers could be identified as being, typically, autosomes from which a substantial part had been deleted, sometimes as the result of a rearrangement with another chromosome. Thus, a karyotype written in the 1960s as 47,+mar might come to be recognized, in the latter twentieth century, as, for example, 47,+der(13), 47,+rea(22), 47,+i(9p),
47,+r(8), or 47,+inv dup(15). With yet subtler ability to dissect out the component parts of these chromosomes, only the much smaller ones remained as a challenge in identification. The expression now mostly in use is “small supernumerary marker chromosome” (sSMC), which Liehr et al. (2004) define thus: “structurally abnormal chromosomes that cannot be identified or characterized unambiguously by conventional banding cytogenetics alone, and generally are equal in size or smaller than a chromosome 20 of the same metaphase spread.” Those supernumerary chromosomes that can be identified and characterized unambiguously, we simply refer to as “der,” according to that identification and character (ISCN, 2009).1 Some sSMCs produce an abnormal phenotype but many, more than half, do not (Liehr et al., 2006). The actual content of the chromosome will determine its pathogenicity or otherwise. Those containing acrocentric short arm and pericentromeric material, or other autosomal pericentromeric chromatin, are typically harmless (Callen et al., 1992). Liehr and colleagues maintain a Web site of sSMCs containing pericentromeric euchromatin, distinguishing those with and without phenotypic consequence (http://www.med.uni-jena.de/fish/sSMC/ 00START.htm). The sSMC with a normal phenotype might nevertheless be the basis for male infertility, as the additional chromosome may interfere with the meiotic process. In a study of 1000 men with nonobstructive infertility, Elghezal et al. (2006) found one man with a supernumerary marker, although not much weight can be put on a single case (see also section on “Details of Meiotic Behavior”). One of the largest family studies concerning an sSMC is described in Bakshi et al. (2008). A 50-yearold woman had her chromosomes tested because she had a tongue cancer, and the entirely fortuitous discovery of a very small marker was made. FISH and whole-chromosome painting showed is to be of chromosome 21 origin. In her family, of 11 normal individuals karyotyped, four had the same sSMC, which was seen, as in her, in mosaic state. This tiny remnant of chromatin had tagged along, transmitted through these several meioses, somewhat incontinently surviving mitosis (that is, generating mosaicisms), and all the while completely harmless.
1 In cancer cytogenetics, the complexities of rearrangement are such that even a large abnormal chromosome might not readily be identified, as to the nature of its component parts. Here, there word marker can continue to be used, whatever the size of the chromosome.
Parental Autosomal Aneuploidy •
239
Crolla (1998) reviewed the published studies of supernumerary chromosomes from 1991 to 1998, numbering some 168 cases. The list is exhaustive, and every chromosome other than chromosome 5 is represented (Masuno et al., 1999, subsequently completed the list with their case of 47,XY,+mar(5)/ 46,XY). Many were associated with phenotypic abnormality, in some severe; some ascertained at prenatal diagnosis eventually proved to be normal. Given the wide range of cytogenetic heterogeneity, of course it is completely unsurprising that there should be a wide phenotypic range, and including normality. Each chromosome needs to be assessed on its merits. As well as the direct analysis of the marker itself, a wider family study can clarify the interpretation. Thus, Takeno et al. (2004) describe a four-generation pedigree, the great-grandmother and the grandmother having the karyotype 46,XX,t(4;13) (p16;q11), with one of the derivative chromosomes, the der(13), being very small. (This chromosome is very similar to the der(13) illustrated in Fig. 5–12). Two sons in the next generation had a tertiary trisomy and a tertiary monosomy, respectively; and the son with the tertiary trisomy, 47,+der(13), then went on to transmit the very small derivative—a chromosome formerly known as “marker”—to his daughter. A supernumerary chromosome that could be pathogenic in nonmosaic form might, in the mosaic state, have no apparent phenotypic consequence. For example, Rothenmund et al. (1997) identified a sSMC in a father as derived from the pericentromeric region of chromosome 8, he having the mosaic karyotype 46,XY[90]/47,XY,+der(8)[10], while his two mentally handicapped daughters had a (nonmosaic) karyotype 47,XX,+der(8). Some particular chromosomes contribute more to the sSMC population than others, one such being chromosome 22. Crolla et al. (1997) propose that der(22) markers can be sorted into those with and those without euchromatin, the former typically being with phenotypic consequence, and the latter not. Urioste et al. (1994b) describe a familial unstable supernumerary chromosome with a mother and two daughters having variable manifestations of cateye syndrome due to 46,N/47,+der(22). Partial autosomal aneuploidy resulting from a ring chromosome, as the supernumerary chromosome, is dealt with in Chapter 11. (This separation into chapters, while pragmatic clinically, may not be
240
•
entirely appropriate, given that many very small markers, subjected to a fine molecular dissection, may turn out actually to be rings; Baldwin et al., 2008.) DE TA I L S O F M EI OT I C B EH AV I O R
The supernumerary chromosome would probably form a univalent at meiosis rather than synapsing with whatever chromosome from which it was derived. Martin et al. (1986a) analyzed sperm chromosomes from two men who had bisatellited markers. Slightly less than half the sperm were found to carry it, although the distribution did not differ significantly from 1:1. However, Cotter et al. (2000) showed a much reduced frequency of sperm carrying a marker, 6% compared with an expected 50%, in a phenotypically normal man with 47,XY, +del(15)(q11.2). Very small markers are prone to loss during cell division. Consequently, mosaicism and, in the case of a transmitted chromosome, familial mosaicism are seen frequently (Chudley et al., 1983). Adhvaryu et al. (1998) describe a bisatellited 15-derived SMC in only 2% of cells in a grandfather, presumed of postzygotic origin in him, whose daughter and a grandson were nonmosaic 47,+SMC, with the chromosome evolving in another grandchild into a very small ring. All these people were phenotypically normal. Familial SMCs are characteristically maternally transmitted, which could reflect preferential exclusion of the marker in spermatogenesis or, in some, a reduced male fertility, as noted earlier. Jaafar et al. (1994) studied a phenotypically normal man presenting with infertility who had a supernumerary bisatellited heterochromatic chromosome. In most spermatocytes, the marker was in close proximity to the X-Y bivalent, and this may have been the cause of the infertility.
GENETIC COUNSELING Parental Trisomy 8 or Trisomy 18 Mosaicism The very rare individual for whom genetic counseling may be a practical issue is likely to have a lowgrade mosaicism, and the diagnosis may have been made because of reproductive loss. Recurrent miscarriage may reflect a higher fraction of trisomic tissue in the gonad, and the risks for a further pregnancy may be substantial. Wei et al. (2000) undertook
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
preimplantation genetic diagnosis in one case, an infertile man with mosaic trisomy 18. A separate issue relates to the possible risk for hematological malignancy in individuals with trisomy 8 mosaicism (Seghezzi et al., 1996; Brady et al., 2000). Trisomy 8 is well known as an acquired change in the cascade of carcinogenesis in myeloid neoplasias (particularly myeloid leukemia, myelodysplastic disease). It may be that constitutional trisomy 8 acts similarly, in this case as the “first hit,” in those individuals in whom the trisomic cell line includes the marrow (as of course it must in patients diagnosed on a peripheral blood karyotype). The level of risk for cancer predisposition is unknown. For the myeloid neoplasias, unlike many other cancers, little therapeutic advantage comes from earlier diagnosis, and the counselor will need to consider carefully how, or if, this issue may be raised (see also p. 15). Parental Trisomy 21 Mosaicism. Theoretically, the risk for having a child with (nonmosaic) Down syndrome is high, up to 50%. Presumably the risk is related to the proportion of gonadal cells that are trisomic, but this is not accessible information. The proportion seen on lymphocyte analysis offers no real help in this question. One point is clear: it is certainly appropriate to offer prenatal diagnosis. Indeed, preimplantation genetic diagnosis may be warranted, as Conn et al. (1999) attempted with a woman who had presented with a history of three out of four pregnancies with trisomy 21, herself with a nonmosaic normal karyotype on blood. Three of four unfertilized oöcytes were hyperhaploid 21, and four out of six analyzable embryos were trisomic 21 (some mosaic). Transfer of the two chromosomally normal embryos was not successful. Parental Down Syndrome. The risk to conceive a trisomic 21 conceptus is presumably 50%, at least in the case of maternal DS. The other parent is likely to have a degree of mental deficiency, possibly due to a genetic factor. Prenatal diagnosis would of course detect those fetuses with trisomy 21, but the risk of a chromosomally normal fetus having a birth defect or being mentally handicapped could be as high as 30% (Shobha Rani et al., 1990). Prevention of pregnancy in those DS women at risk is regarded
by many as advisable. In two confirmed cases of paternity in a DS man, prenatal diagnosis (with a normal result in both) was performed (Bobrow et al., 1992; Pradhan et al., 2006). We refer the reader to the discussion in Chapter 1 (p. 17) for a general review of the ethical issues in this setting. Parental Partial Autosomal Trisomy or Monosomy. The risk for a child to have the same defect as the parent is 50%, or very close to it. A lesser risk applies in the case of mosaicism. In the case of a person of only low-normal/borderline intelligence who is functionally fertile, the issue of the risk and possible questions of prenatal diagnosis or sterilization will be difficult to raise (p. 17). A phenotypically normal individual said to have a partial aneuploidy merits a cytogenetic or microarray reevaluation, to check for the possibility that the supposed aneuploidy is actually a balanced rearrangement that was only partially characterized. As cytogenetic techniques have become refined, new “defects” are being identified which, upon family study, are revealed as being unusual, but functionally balanced, forms (Barber, 2005). For practical purposes, they can be regarded as variants, rare or possibly even “private” to that family. The counselor must take care that apparently abnormal karyotypes are not overinterpreted. In a family study undertaken to clarify a particular unusual chromosome, family members should understand the reason for the study. The Harmless Supernumerary Marker Chromosome. Each chromosome needs to be judged on its merits, a detailed cytogenetic assessment and parental karyotyping study having been undertaken. If a SMC is judged to be harmless, then on that basis it would be immaterial whether a parent transmits it to an offspring. Nevertheless, it may be useful to know that, in a particular family, there has not been any associated phenotypic abnormality, a point that may be settled by doing a family study; this enables the counselor to offer firm reassurance that those who inherit it in the future will also likely be normal. A possible association with male infertility is mentioned earlier. The very rare circumstance of an sSMC identified in a normal person—and seeming, at first sight, harmless, but the sSMC then revealed to have been derived from a deletion on the homolog concerned— is dealt with on p. 158. These carriers have a high risk for abnormal offspring.
Parental Autosomal Aneuploidy •
241
15 the fragile x syndromes
FRAGILE XA SYNDROME appeared on the cytogenetic scene in the late 1970s, although by the mid-1990s it occupied a predominantly molecular genetic stage. But it is entirely appropriate that it retains a place here, and indeed a whole chapter. Fragile XA syndrome (or simply fragile X syndrome; FXS, in common parlance) is the second most prevalent genetic cause of mental deficiency after Down syndrome, and it is the most common familial cause. The premutational state is associated with a degenerative neurological syndrome typically of onset in late middle age, and, in the female, ovarian insufficiency; and we may speak of the fragile X associated syndromes (FXSs). Fragile XE syndrome is rare, but it is one of the more common forms of nondysmorphic mild intellectual disability.
BIOLOGY Fragile XA syndrome (FXS) is named for the folatesensitive fragile site, FRAXA, at Xq27.3 (Fig. 15–1). The first family in which this fragile site was reported
242
•
was seen, initially, as a fascinating story but of limited practical relevance (Lubs, 1969); some four decades later, the FXDs command a prominent stage worldwide. There are two other rare fragile sites distal to FRAXA, named FRAXE and FRAXF. FRAXE is a rare folate-sensitive fragile site that is the chromosomal manifestation of an expanded CCG repeat that silences the FMR2 gene, which is the molecular basis of its phenotype of nonspecific mental retardation (Gecz et al., 1996). FRAXF appears to be a harmless rare variant, although the full mutation does silence the FAM11A gene (Shaw et al., 2002). FRAXD is a common fragile site, proximal to FRAXA, and it is also harmless and a part of normal chromosome structure. The FRAXA fragile site exists within the 5′-UTR of the FMR1 (fragile X mental retardation-1) locus, which encodes a protein named FMRP (fragile X mental retardation protein). FMRP acts as a regulator of mRNA transport and of translation of specific mRNAs at the synapse (Oostra and Willemsen, 2009), and it is necessary for normal brain development
(a)
(b)
(c)
(d)
FIGURE 15–1 Plain-stained sex chromosomes from a normal female (a) and male (b) compared with those of a fragile X female (c) and a fragile X male (d). The fragile site is arrowed. The segment distal to the fragile site appears as a satellite, as though it is about to break off.
and function. The nature of the mutation in FXS is an expansion of a normal sequence of trinucleotide (CCG) repeats within this 5´ region. The different fragile X trinucleotide repeats (FMR1 alleles) can be grouped into four size categories: common (6–40 repeats), intermediate or gray-zone (41–58 repeats), premutation (59–200 repeats), and full mutation (>200 repeats). The classic clinical picture of FXS due to the full mutation will be familiar to most counselors: intellectual disability, mood disturbance, and abnormal behavior that may merge into frank autism, and physical traits including a subtly distinctive facies, joint hypermobility, and, in the male, macro-orchidism. A child from the original family reported by Lubs (1969) is shown in Figure 15–2. I NCI DEN CE
The full mutation exists in 1 in 4000–5000 males, as measured from indirect and direct approaches, an example of the latter being a study of 36,124 newborn males having routine bloodspot screening (Crawford et al., 2001; Coffee et al., 2009). An important figure
FIGURE 15–2 A child with fragile X syndrome from the family in which the fragile site was first recognized. The facies is characteristic. (From H. A. Lubs, 1969, A marker X chromosome, American Journal of Human Genetics 1969;21:231–244. Courtesy H. A. Lubs; reproduced with the permission of the American Society of Human Genetics.)
is the frequency of the female premutation carrier, these women being at risk to have a child with FXS. Somewhat different figures have been derived from different populations. In Quebec, about 1 in 260 women carry a premutation of 55–101 copies of the CCG repeat (Rousseau et al., 1995), while in Finland the fraction is very similar at 1 in 250, for women having more than 60 copies of the repeat (Ryynänen et al., 1999). In Israel, the figure is higher, and in Japan, less. One in 150 Israeli women have 55 or more repeats (Berkenstadt et al., 2007), while in Japan the incidence of fragile X appears to be lower The Fragile X Syndromes •
243
than in other populations, and Otsuka et al.(2010) found no premutation carriers among 576 men and 370 women, albeit that these denominators were rather small.1 In theory, the male premutation carrier prevalence should be close to one half of the female rate. One estimate is 1 in 1000 (Crawford et al., 2001), although this figure is to be seen alongside the 1 in 250 fraction that Fernandez-Carvajal et al. (2009b) derived from a study of 5267 male newborns having blood spot screening in northwestern Spain. For the record, in this Spanish study, the frequency of the intermediate (gray-zone) repeat in the male was 1 in 26. Castellví-Bel et al. (2000) showed that the incidence of intermediate FMR1 alleles was not significantly different in retarded and in appropriate control populations, although recent suggestions of a possible morbidity association are noted later. Intellectual disability associated with FRAXE is rare and probably affects less than about 1 in 100,000 individuals (Gecz, 2000). Sporadic patients with FRAXE may not be routinely diagnosed, testing only being performed when there is a family history of intellectual disability under investigation.
Molecular Genetics of the Fragile X Loci FRAX A (FMR 1 )
Fragile XA syndrome is a “dynamic mutation” disorder (Sutherland and Richards, 1993). As outlined
earlier, there is a section of DNA at the fragile site comprising the triplet cytosine-cytosine-guanosine (CCG) repeated many times.2 The number of repeats determines the genotype of normality, “gray zone,” premutation, or full mutation (Table 15–1). Normal X chromosomes have from 6 to about 58 sequentially repeated copies of the CCG triplet, with copy numbers between 40 and 58 being regarded as in a “gray zone.” Hemizygous carrier males with the premutation have approximately 58–200 copies of the triplet-repeat, as do female heterozygotes on one of their X chromosomes. Males with the “full mutation” of more than 200– 220 triplet-repeats (up to about 1000 copies) have FXS. The situation is a little more complex for females with the full mutation, reflecting variable Lyonization in the brain, with some neurons expressing the normal, and some the abnormal chromosome. Of female carriers with the full mutation, about half have some degree of intellectual disability, but rarely of a similar degree to the male (Thompson et al., 1994). The CCG triplet-repeat sequence, when increased beyond a critical size (in the vicinity of 55 copies), is unstable and can change in copy number when transmitted from parent to child. Thus, a mother with a premutation can transmit a full mutation to her child. A mother with a full mutation can transmit a larger or smaller full mutation to her child. A mother with a “gray zone” allele can transmit an expansion of this allele to her child, into the
Table 15–1. Trinucleotide Lengths and Associated Phenotypes NO. CCG REPEATS GROUP
MALE COGNITIVE PHENOTYPE
FEMALE COGNITIVE PHENOTYPE
6–39 40–54 55–200 >200
Normal Normal Normala Mentally retarded
Normal Normal Normalb Variable
Normal “Gray zone” Premutation Full mutation
a
Possible cognitive impairment and fragile X tremor/ataxia syndrome in later middle age (see text and Table 15–2). Some may show a mild psychological/personality effect; and there is an association with premature ovarian failure and fragile X tremor/ataxia syndrome (see text and Table 15–2). b
1 There is precedent for triplet-expansion disorders having variable population frequencies. For example, the relative frequencies of Huntington disease and the somewhat similar neurodegenerative disease DRPLA (dentato-rubro-pallido-luysian atrophy) are the reverse in Japan, compared with European populations: HD is very rare in Japan, and DRPLA very rare in Europeans. 2 There is some confusion in the way trinucleotide repeats are expressed (Sutherland and Richards, 1993). Taking the convention that the bases be written in alphabetical order from 5´ to 3´, the FRAXA repeat is CCG. Some authors “read” the opposite DNA strand, and write CGG instead (or AGG in place of CCT).
244
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
premutation range, but not as a full mutation. In contrast, fathers only transmit premutations to their children (necessarily only their daughters) regardless of whether they themselves carry a premutation or, very rarely, a full mutation. (While very rare exceptions to this have been reported, e.g., Zeesman et al., 2004, they can usually be disregarded in practice.) The change from normal copy number to full mutation is a multistep process, proceeding through premutation steps presumably over a number of generations, rather than as a single event characteristic of classical mutation. The likelihood of expansion is related to the presence of “AGG interruptions” (or CCT2 interruptions) in the premutation repeat: the longer the run of pure CCGs, the greater the propensity for expansion (Nolin et al., 2011). Laboratory Analysis. The varying sizes of the fragile site DNA can be visualized on a Southern blot. Figure 15–3 shows the patterns seen in various types of individual and demonstrates the instability in a family, and further examples are shown in Figure 15–4. If DNA is digested with the enzyme PstI and probed with pfxa3, the normal X chromosome gives a fragment of approximately 1.0 kb. This ~1.0 kb band represents about 900 bp of DNA flanking the repeat plus the 18–165 bp of the actual CCG repeat sequence itself (which amounts to 6–55 tripletrepeats). The DNA fragment from a fragile XA chromosome, having within it the additional copies of the CCG repeat sequence, is larger by this amount. For example, in a person with 200 copies (200 × 3 = 600 bp) of the triplet-repeat—which is about where the premutation merges into the full mutation—the fragment is ~1.6 kb (~1000 bp + 600 bp) in size. This is an increase (“amplification”) of ~600 bp over the normal size of 1 kb. Basehore and Friez (2009) outline a protocol for the complementary application of polymerase chain reaction (PCR) and methylation-sensitive Southern analysis. FUL L M UTATIO N : MET HY LAT I ON OF TH E FMR1 G EN E
Hypermethylation has a central role in the mechanism by which the large-size CCG repeat (>200 repeats) causes the molecular pathology that, in turn, leads to the FXS phenotype. The repeat sequence is located within a noncoding portion (the 5’-untranslated region) of the FMR1 gene. Once there are more than about 200 copies of the repeat,
3.60 2.80 1.95 1.86 1.55 1.39 1.16 0.98 0.72
pS8
FIGURE 15–3 Inheritance of the fragile XA unstable element in a four generation lineage from a large affected pedigree. Pedigree symbols: normal carrier female expressing the fragile XA on cytogenetic study = ; affected FXS male expressing the fragile XA = ; normal female = . All carriers, . and , . are obligate carriers. Chromosomal DNA was digested with PstI and probed with pfxa3. The control probe pS8 was included in the hybridization.
the DNA surrounding the repeat, and the repeat itself, become hypermethylated. (Methylation is the addition of methyl groups to the C (cytosine) bases in CpG sequences in the DNA molecule.) Hypermethylation is associated with inactivation, in cis, of the FMR1 gene containing the triplet-repeat sequence, and it ceases to be transcribed: a “loss-offunction” mutation. The consequential lack of the FMR1 gene product (FMRP) is the cause of the abnormal phenotype of the FXS. The normal function of FMRP is to control the activity of proteins
The Fragile X Syndromes •
245
1
2
3
4 5
6
7 8
9
pf x a3 1.0
pS8
0.8
FIGURE 15–4 Southern blot characteristics of different genotypes. (1) Carrier female, (2) transmitting male, (3) noncarrier female, (4) affected male, (5) carrier female, (6) affected male, (7) noncarrier female, (8) normal male, and (9) carrier female. PstI-digested DNA probed with pfxa3 and pS8.
involved in the pre- and postsynaptic machinery of the central nervous system; absence of FMRP leads to dysregulation of these proteins, and thus compromise of brain functioning.3 A specific observation in brain tissue from patients with FXS is an excess of immature forms of the “spines” that project from the branches (dendrites) of the neurons (Irwin et al., 2001), and which comprise the physical basis of synapses (connections between neurons). Particular regions of the brain may be susceptible, and Haas et al. (2009), using sophisticated imaging analyses, point to certain white matter tracts that connect the left frontal lobe to deep structures (the ventral frontostriatal pathway), and which contribute to the control of appropriate inhibitory behavior, as having an abnormal fiber density. Determination of repeat copy number and, if necessary, assessment of methylation as an indicator
of the activity state of the gene, can be used to help predict phenotype. Methylation status of the tripletrepeat and adjacent DNA in affected males cannot be resolved from the routine FMR1 PstI digestion DNA test (see later discussion). “High-functioning” affected males may have amplification values of between 0.6 and 1.0 kb above the normal size. If a more accurate genotype-phenotype correlation is required, analysis of methylation status is done by probing a double digest of EcoRI and EagI. EcoRI/EagI double digests of DNA from males within this range of amplification values and probed with pfxa3 are shown in Figure 15–5 (the probes StB12.3 and Ox1.9 would give an identical result). Interpretation is simpler in males than in females. The normal chromosome of carrier females is also methylated in half the cells, on average, as part of the normal random X-inactivation process. While methylation may have a role, it is not the only process influencing the function of FMR1. In the absence of methylation, the fully expanded (>200 repeats) gene is overtranscribed, but the mRNA is inefficiently translated, and the degree of inefficiency increases in proportion to repeat copy number. Feng et al. (1995) analyzed clones of cells having 207, 266, and 285 repeats: these produced, respectively, 24%, 12%, and essentially 0% of the normal amount of FMRP. Possibly, this reduction in protein is cell type specific and certain tissues (for example, chorionic villi) with large unmethylated repeats can still make some FMRP. PREM UTAT I O N : R N A TO XI CI TY
The FMR1 premutation4 has its pathogenic effect due to an entirely different mechanism: a toxic gainof-function. The expanded (55–200 repeats) FMR1 gene is transcribed, and in increased amounts; and these enlarged RNA transcripts then behave abnormally. They aggregate within the nucleus of the neuron, and these aggregations attract certain other species (e.g., lamin A/C, αB-crystallin). These several substances are sequestered as intranuclear inclusions. Over time, these inclusions compromise the functioning of the neuron, and thus the functioning of
3 A very small fraction of FXS boys look rather as though they have Prader-Willi syndrome. This effect might be a consequence of a lack of FMRP leading to an inadequate production of CYFIP (cytoplasmic interacting FMRP protein), this protein having its gene within the PWS region of chromosome 15 (Nowicki et al., 2007). 4 The word premutation is well entrenched in fragile X jargon, but it betrays its etymology from when a transmitted X not displaying the fragile site was considered to be, in the 1980s, simply a state from which the true mutation might then arise. Now, as a bona fide mutation in its own right, it is stuck with this somewhat inaccurate nomenclature.
246
•
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
1
2
3
4
5
6
7
8
9
10
11 12
5.2kb
2.8kb
FIGURE 15–5 Restriction patterns for fragile X syndrome males who show methylation differences. EcoRI digests are shown in odd-numbered lanes; EcoRI/EagI double digests are shown in even-numbered lanes. Normal control male, lanes 1–2 and 11–12. The normal EcoRI fragment is 5.2 kb and the normal unmethylated EcoRI/EagI fragment is 2.8 kb. Lanes 3 through 10 for affected individuals show fragments of higher-thannormal molecular weight because of amplification. (Females would exhibit additional complexity because additional methylated and unmethylated bands occur for the normal X chromosome.) The affected individual in lanes 3–4 is unmethylated. Lanes 5–6 is a methylation mosaic; most of his cells have the EagI restriction site unmethylated, but in some cells it is methylated. The individual in lanes 7–8 is fully methylated, and the one in lanes 9–10 is unmethylated. The tissue tested was blood, which may not necessarily reflect methylation patterns in other tissues. (From D. Z. Loesch et al., 1993, Genotype-phenotype relationships in fragile X syndrome: a family study, American Journal of Human Genetics 53:1064–1073. Reproduced with the permission of the American Society of Human Genetics.)
the various component parts of the neural substrate in which this process is taking place (Greco et al., 2002; Brouwer et al., 2009; Gokden et al., 2009). Some other classes of cell are also affected, for example, the hormone-producing cells of the testis and thyroid; and a similar process affecting the ovary is likely the basis of the premature ovarian failure in the female ( Jacquemont et al., 2007).
The Effect of FMR1 Premutations on Carrier Males Neuropsychological Capacity Many men may be free of neuropsychological complication (Hunter et al., 2010), but a few children with high-end premutations may manifest learning difficulty. Some
premutation males may display deficits of executive functioning and memory on neuropsychological examination (Moore et al., 2004). Men over 50 with larger (70–200) repeats have a six-fold increased risk for a progressive cognitive impairment (Sévin et al., 2009). The Tremor-Ataxia Syndrome. The FMR1 premutation can lead to a neurodegenerative disorder, first described in 2001: the “fragile X tremor/ataxia syndrome” (FXTAS) (Hagerman et al., 2001; Brouwer et al., 2009). The most severe involvement is expressed as a progressive neurological syndrome with cerebellar and parkinsonian features, accompanied by decline in executive function and cognitive capacity, with overt onset usually in late middle age. The Fragile X Syndromes •
247
A consistent neuropathological pattern, with widespread neuronal intranuclear inclusions, has been shown at postmortem. The cerebellar neuroradiology is characteristic and may precede actual onset of symptoms (Brunberg et al., 2002; Loesch et al., 2008). The penetrance of FXTAS is in the range of 33%–45%, and thus about a third to almost a half of premutation males will develop this condition, in an age-dependent and repeat size-dependent manner ( Jacquemont et al., 2004; Rodriguez-Revenga et al., 2009). The smallest premutation to result in FXTAS is 69 repeat copies. The risk in Sévin et al. (2009) was greater (33%) for midsize/large (70–200 CCG) repeats than for smaller ones (55–59 CCG), the penetrance in the latter group of 5.9% being close to the 5.1% control figure; this study also noted that cognitive impairment preceded the onset of the actual syndrome. Penetrance estimates are set out in Table 15–2.
The Effect of FMR1 Premutations on Carrier Females Neuropsychological Capacity. Typically, females who carry FMR1 premutations have intelligence within the normal range, but some lines of evidence point to subtle effects, and sometimes more overt psychopathology, in some carriers (Bourgeois et al.,
Table 15–2. Penetrance of Fragile X Tremor/Ataxia Syndrome in Premutation Carriers, Shown as Percentage According to Age AGE (YEARS)
SERIES
JACQUEMONT ET AL. (2004)
50–59 60–69 70–79 > 80
RODRIGUEZ-REVENGA ET AL. (2009)
MEN
MEN
WOMEN
17% (12) 38% (13) 47% (17) 75% (4)
0% (1) 53% (17) 53% (15) 27% (11)
4% (27) 20% (15) 26% (19) 21% (24)
Note: The reduction in penetrance in the >80 group in Rodriguez-Revenga et al. likely reflects increased mortality in the affected individuals. Numbers of subjects shown in parentheses.
248
•
2009). Depression is more frequent, and may be more marked with larger repeats, above 100 CCG copies; it is controversial whether this association might have an intrinsic neurological basis or be secondary to ovarian hypofunction (Hunter et al., 2010). Migraine is common. The Tremor-Ataxia Syndrome. In the initial study of Jacquemont et al. (2004), the incidence of FXTAS symptoms in 59 premutation women was not greater than in controls. However, the same group (Coffey et al., 2008) subsequently found a penetrance of 8% in 72 premutation carriers aged greater than 40 years. Rodriguez-Revenga et al. (2009) showed that 16% of such women aged 50 or more years (rising to 22% of those over 60 years) had signs of FXTAS. Ovarian Insufficiency. Ovarian function is affected, with consequential primary ovarian insufficiency/senescence (POI/S), which may then proceed to actual premature ovarian failure (POF). Allingham-Hawkins et al. (1999) showed that among 395 premutation carriers, 16% had early (before age 40) menopause, compared with only 0.4% in a control group comprising noncarrier relatives. Hundscheid et al. (2000) give likelihoods for POF according to age as follows: 4% by age 30 years, and 25% by age 40 years. The risk for POI/S and for POF correlates with the number of CCG repeats (Sullivan et al., 2005; Hunter et al., 2008b), and a risk for POI/S may also apply to women carrying a gray zone allele, and even an upper-range normal allele (Gleicher et al., 2009; Streuli et al., 2009). Other Complications. More broadly, the carrier state in the female is associated with a range of other medical problems, including in particular thyroid disease and fibromyalgia (penetrances of 16% and 24%, respectively), and with seizures, high blood pressure, and peripheral neuropathy also recorded (Coffey et al., 2008; Rodriguez-Revenga et al., 2009; Chonchaiya et al., 2010). I NT ER M ED I AT E (“ G R AY-ZO N E” ) A L L EL ES
To add further complication, triplet expansions intermediate in size between the accepted “common” range (up to ~40 repeats) and the onset of premutations (~58 repeats) might possibly convey a phenotype, although this is controversial. As noted earlier, premature ovarian insufficiency/senescence may
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
develop in the female heterozygote for a gray-zone allele. A case for cognitive or behavioral compromise, which may present as autism, in a child of either sex, has been proposed (Loesch et al., 2007). Likewise a suggestion has been made of an increased risk for parkinsonism (Loesch et al., 2009; Zhang et al., 2009b), but this remains to be confirmed (Kurz et al., 2007). A single instance of expansion from a gray zone allele to a full expansion is on record: from grandfather (52 repeats) through his daughter (56 repeats) to his severely retarded grandson (~538 repeats); the grandfather displayed tremor and ataxia (Fernandez-Carvajal et al., 2009a). This is the first report of an allele of <59 repeats expanding to a full mutation in a single step. Two Types of FMR1 Mosaicism. Individuals with full mutations can show somatic instability of the amplified repeat sequence. Different cells in a single tissue can be genetically different, in terms of triplet-repeat length; and genetic differences can exist between tissues. This is manifest as a smear of DNA fragments on Southern blot (Fig. 15–4, lanes 4–6). This instability can lead to two types of mosaicism. While strictly speaking individuals with smears of DNA in the full mutation size range are mosaics (there are different lengths of triplet-repeats), this term is reserved for two specific situations. First, in mutational mosaicism there are some cells with full mutations, which are fully methylated; and some cells with premutations, which are unmethylated and the gene can function. The mental phenotype can vary, presumably depending upon the type of mutation predominating in different parts of the brain, and thus the regional activity within cerebral tissues of the FMR1 gene. Up to 20% of FXS males are mutational mosaics detectable on Southern analysis and some may have an IQ within, but at the lower end of, the normal range. A rare type of mutational mosaic is the individual with some cells containing a normal number of repeat copies and others containing premutation or full mutation copy numbers. Polymerase chain reaction is a more subtle tool and may indicate the presence of a premutation in a very small fraction of cells in many FXS males. De Graaff et al. (1995) could show on brain tissue from an affected male that about 1% of neurons expressed FMRP, and thus presumed that these individual neurons had a genetically active premutation. Second, in methylation mosaicism, the number of repeat copies is characteristic of a full mutation, but
the DNA is not methylated in all cells (Fig. 15–5, lane 6). This is less common than mutational mosaicism and occurs mostly at the lower end of the range of full mutation copy number. “Highfunctioning” FXS males may be methylation mosaics, with full mutations which are partly or completely unmethylated. Other Mutational Bases of FRAXA. Almost all FXS is due to triplet-repeat expansion. A very few affected persons have a different mutational basis, and deletions (ranging in size from 1 bp to about 100 bp and up to megabase size) and point mutations in the FMR1 gene have been reported (Hammond et al., 1997; Grønskov et al., 2011). Duplication of a 5 Mb segment within Xq27.3q28 and which includes the FMR1 locus, as well as FMR2 and 26 other genes, leads to a heritable syndrome of mental retardation and short stature with hypogonadism (Rio et al., 2010). These authors propose that a functional disomy of FMR1 contributes to the clinical phenotype. Grønskov et al. (1998) and Castellví-Bel et al. (1999) screened a number of patients with a high clinical score for the features of FXS, and yet found no mutations.
Fragile XE The mechanism is again a CCG repeat that expands through premutations to full mutations (Mulley et al., 1995). The CCG repeat is in the 5´ untranslated region of a gene called FMR2, whose 9.5 kb transcript encodes a putative 1302 amino acid protein. FMR2 transcription is silenced in males with CCG expansion with methylation of the adjacent CpG island (Gecz et al., 1996). Because of its milder phenotype, and because it is so rare, the diagnosis is not often clinically suspected, although it is one of the more common forms of nonsyndromal mild X-linked mental retardation. In some FRAXE families there will be males who have full mutations who are not intellectually disabled, as the cognitive impairment associated with FRAXE overlaps the normal range (Gecz 2000). Microdeletion within the FRAXE gene has been identified on microarray (Sahoo et al., unpublished data), and it may be that further such cases will be recognized with the increasing use of molecular methodology. Fragile XF. FRAXF is also due to dynamic mutation of a CCG repeat (Parrish et al., 1994), but in this case an apparently harmless one, in that it is The Fragile X Syndromes •
249
not associated with a familial form of intellectual disability. CYTOGEN ETICS
The appearance of the X chromosome in FXS is now of essentially academic interest. But historically, this is what gave the syndrome its name, and the counselor should be aware of this history. The “fragility” refers to a fragile site on the chromosome at which the chromatin is attenuated, and which resembles a gap or break (although there is in fact usually continuity) (Fig. 15–1). In the early days of the syndrome, laboratory technology was developed that stressed the chromosomes, and this was essential for the fragile site to manifest itself and to be seen by the cytogeneticist (Sutherland, 1991). These induction methods all led to a relative deficiency of either thymidine or deoxycytidine at the time of DNA synthesis. It is of interest that the fragile site was seen in almost all affected FXS males and most females, and in most carrier females who had the full mutation, but not in carriers (male or female) with a premutation. As well as the FRAXA fragile site, other sites within distal Xq are FRAXE, and the normal variants, FRAXD and FRAXF (see earlier).
Genetics FMR1
The inheritance of FXS is certainly X linked, but it becomes a matter of semantics as to whether this might be “X-linked semidominant,” or “X-linked incomplete recessive.” More usefully, we may refer to the premutational or full mutational state, to the associated gender-specific penetrances, and to the probability of expansion from the premutational to full mutational state upon transmission of the chromosome to a child. The following points may be noted. De Novo Full Mutations Do Not Happen No new mutation has been observed; that is, no individual with a full mutation has been observed as the offspring of parents who have normal numbers of copies of the CCG repeat on their X chromosomes. The mothers of all FXS individuals are carriers of at least a premutation, and where study has been possible, so is a grandparent. The rate of mutation from normal CCG copy number to premutation thus appears to be very low. 250
•
Male Transmission. When the unstable sequence is transmitted by males, it characteristically does not increase in size, and it may decrease. In males with full mutations, only premutations are seen in sperm (Reyniers et al., 1993). (There are extremely rare exceptions to this rule: Zeesman et al. (2004) reported a male who was a premutation/full mutation mosaic whose daughter was also a premutation/full mutation mosaic.) Female Transmission. When the unstable triplet-repeat sequence is transmitted by females, it usually increases in size (although rare decreases have been reported, and even more rarely, gene conversion events have reduced mutation-range fragile X chromosomes back to normal; Losekoot et al., 1997). Women who transmit X chromosomes with small premutations usually show less increase in size than women who transmit larger premutations. Thus, women with fewer than 70 triplet-repeats (“low-end” premutations) mostly have children who also have premutations, although the premutations in these sons and daughters are characteristically larger than those of their mothers. On the other hand, women with “highend” premutations (90 or more triplet-repeats), and carriers who themselves have a full mutation, almost always transmit a full mutation (Table 15–3). The risk is influenced by whether there are “AGG interruptions” in the premutation repeat sequence (see earlier), and this may be the molecular basis of the observation that the risk for expansion is greater in those women who have a known family history of FXS, versus those ascertained by other means (e.g., through a screening program) (Nolin et al., 2011). F RA XE ( FMR2 )
Not enough fragile XE families have been reported in detail for segregation analyses to be performed. Notably, and distinct from FRAXA, instances of a FRAXE full mutation being passed from father to daughter are on record (Hamel et al., 1994; Carbonell et al., 1996). RA R E CO M PL EXI T I ES
Bringing together the two Fragile X syndromes, we have seen a family in which both FRAXA and FRAXE are segregating (Mulley et al., 1995). A FRAXE man married a FRAXA heterozygote,
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
Table 15–3. Risks for Fragile X Premutation Carriers to Have a Child with a Full Mutation, If the Fragile X Chromosome Is Transmitted PREMUTATION OFFSPRING SIZE (TRIPLET- WITH FULL MUTATION/ REPEATS, n) ALL FRA(X) PREGNANCIES
RISK FOR FULL MUTATION IF FRA(X) TRANSMITTED (%)
Nolin et al. (2003) 55–59 1/27 60–69 6/113 70–79 28/90 80–89 81/140 90–99 89/111 100+ 194/197
4 5 31 58 80 98
Berkenstadt et al. (2007) 55–59 0/79 60–69 3/77 70–79 4/9 80–89 6/11 90–99 2/2 100+ 13/13
0 4 44 55 100 100
Nolin et al. (2011) 45–49 0/55 50–54 0/51 55–59 0/86 60–69 2/81 70–79 15/47 80–89 45/61 90–99 31/33 100+ 93/95
0 0 0 2 32 (54%; 11%)* 74 (88%; 33%)* 94 98
Note: The risk becomes higher with a larger expansion size in the mother; the risks are less for mothers ascertained other than through having a family history of fragile X syndrome. *The % figures in parentheses for allele sizes 70–79 and 80–89 from Nolin et al. (2011) show the higher risks for mothers with a family history of fragile X syndrome (54%, 88%), versus lower risks for women ascertained otherwise (11%, 33%). Sources: Data are from three large studies: Nolin et al. (2003), Berkenstadt et al. (2007), and Nolin et al. (2011). The former group was ascertained through an affected fragile X syndrome individual, and the risk figures appropriately adjusted; the latter two are based upon prenatal diagnostic data. In Berkenstadt et al. (2007), the women were recruited prospectively as part of a broad screening program; in Nolin et al. (2011), a retrospective analysis was undertaken from multicenter data.
and they had two daughters, one a FRAXE heterozygote and the other a FRAXA/FRAXE compound heterozygote. The latter, in turn, has had a son with FXS, and FMR2 and FMR1 carrier daughters. There are cases reported of women who are FMR1 compound heterozygotes, having a full mutation on their maternal X, and a premutation on the X from their father (Linden et al., 1999; Hegde et al., 2001). The fragile X phenotype can coexist with other abnormalities as part of a contiguous gene syndrome, or simply by chance. Quan et al. (1995) report a child in whom a deletion in Xq26.3–27.3 removed the FRM1 and adjacent loci. His phenotypically normal mother showed selective inactivation of the deleted X. A mother described in Missirian et al. (2000) was heterozygous for an FMR1 premutation and a 22q11.2 deletion. Two of her children had DiGeorge syndrome (one also with a FMR1 premutation), and one had FXS with a full mutation. Diagnosis. As outlined earlier, laboratory investigation is typically based upon PCR amplification of the CCG repeat and sizing by capillary electrophoresis. This can be used for both FMR1 and FMR2. Males without a clear product in the normal size range, and females who are not clearly heterozygous for two normal alleles, should then be studied by Southern blot analysis for amplification of the repeat. For prenatal diagnosis, Southern blot is the gold standard, when PCR cannot demonstrate normal parental allele(s) in the fetus. Screening for Carrier Status in Women of Reproductive Age. The prevention of FXS, in further pregnancies in a family where an affected child has been identified, is “secondary prevention.” Primary prevention would require that carrier women be detected ahead of having a child with fragile X: in other words, in families without a proband. Toledano-Alhadef et al. (2001) describe their experience of offering screening to women in Israel. They tested 14,334 women without any family history of mental retardation and found 3 full mutation carriers and 127 premutations of 55 repeat copies or more. Among the premutation carriers who were, or who became pregnant, prenatal diagnoses were performed in 97, and 5 full mutation fetuses were detected. This small risk for a full mutation (only 5%) may reflect different likelihoods for premutations to expand to a full mutation in cases ascertained other than through an FXS proband (compare the The Fragile X Syndromes •
251
two groups listed in Table 15–3). Alternatively, most of the premutations in this group may have been right at the low end of the CCG premutation range. These findings were confirmed by Berkenstadt et al. (2007) in the Israeli population where ~1 in 150 women were identified as premutation carriers. They found that the incidence of premutations in women with a family history of mental retardation was the same as that in women without such a history, but the size of the premutations in the former group was larger and thus resulted in a higher risk of expansion to full mutations. Decisions to conduct such screening programs will depend upon funding of health care locally, and may be influenced by the frequency of carriers in the population (which is apparently higher in Israel than in other comparable populations). In a detailed economic analysis, based on the particular circumstances obtaining in The Netherlands, Wildhagen et al. (1998) determined that the cost of detecting one carrier is about US$45,000, whether testing be conducted through prenatal clinics, by preconceptual screening, or through schools. This figure contrasts with lifetime costs of care for FXS males and females of about $960,000 and $530,000, respectively. Time will have changed these amounts, but their relativity is likely similar. A separate question is the testing of newborns for full FMR1 mutations, which could, in principle, readily be added to an existing newborn screening program, with dried blood spots used for an assay of FMR1 methylation. Potential benefits from early diagnosis would be to enable timely genetic counseling, ahead of the parents having subsequent children, and of earlier clinical intervention (Coffee et al., 2009).
GENETIC COUNSELING As always, good genetic counseling depends upon accurate diagnosis. In the context of fragile XA, this means the molecular confirmation of the diagnosis of FXS and the molecular identification of carriers with good estimates of the number of repeat copies they carry. As McConkie-Rosell et al. (1999) point out, women who are carriers often have strong views about the desirability of avoiding having affected children, and want their relatives to be informed. People have different coping mechanisms to deal with the challenge of learning of their carrier status, and McConkie-Rosell et al. (2001) reproduce excerpts from a number of carriers they had 252
•
interviewed, illustrating this fact. These authors have also addressed the issue of what might be suitable ages to discuss the question of the genetic risk, and actually to offer testing: each individual needs to be judged on his or her merits, and “there is no universal ‘right’ age” (McConkie-Rosell et al., 2002). Consensus statements on genetic counseling for fragile X have been issued by a number of groups, and the counselor should be familiar with these (McConkie-Rosell et al., 2005; American College of Obstetricians and Gynecologists Committee on Genetics, 2006; Chitayat et al., 2008, for the Society of Obstetricians and Gynaecologists of Canada, and the Prenatal Diagnosis Committee of the Canadian College of Medical Geneticists). The picture in FRAXE is less clear; FRAXD and FRAXF are not of practical concern.
Risks of Having an Abnormal Child F RA XA ( FMR1 )
Male with Premutation. For carrier males (“normal transmitting males”) there is, according to current knowledge, virtually no risk to have a mentally impaired child due to the FRAXA gene per se. All daughters receive the FMR1 gene in its premutation form, whatever its size in terms of tripletrepeats. It is practically unknown for a daughter to receive a full mutation. Thus, practically invariably, none would have FXS. All 46,XY sons receive their father’s Y chromosome, and obviously his carrier state implies no genetic risk to them. The extremely rare instance of having a daughter with a full mutation could be mentioned, but the very low probability of this happening should be stressed (Zeesman et al., 2004). Male with Full Mutation. Procreation in this group is extremely rarely documented. Retrogression to a premutation in sperm dictates that daughters would be premutation carriers and (other things being equal) of normal intelligence. Sons receive the Y chromosome. There may be extremely rare exceptions to this, with a daughter receiving a full mutation (Zeesman et al., 2004). Female with Premutation. The risk of transmitting the FRAXA X chromosome is the typical mendelian 50%. If it is transmitted, the risk of having a child with FXS (in other words, a child with a full
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
mutation) depends upon the size of the mother’s premutation (Table 15–3), and upon the way the family came to attention. Women with “low-end” premutations of less than 60 copies of the tripletrepeat have only a very small risk to have a child with FXS; 56 copies is the smallest premutation reported to have expanded to a full mutation in one generation, from a single report (Fernandez-Carvajal et al., 2009a), as noted earlier. At the other end, for those with more than 90 copies (“high-end premutation”), the risk is in the range of 90% to 100%. The risks are substantially less in women who have been ascertained other than through a family history of FXS (Nolin et al., 2011). There is the additional concern of the neurodegenerative syndrome of FXTAS coming on in late middle age in some who inherit a premutation (Table 15–2). Ovarian Function. The risk for primary ovarian insufficiency/senescence (POI/S) has been noted earlier. Women with approximately 80–100 repeats are at greatest risk, with one-third experiencing premature ovarian failure (POF), although only about 4% before age 30 years. These risks are lower for those with both smaller and larger premutations. Family genetic background appears to influence the risk, and a family history of POF points to an increased likelihood (Hunter et al., 2008). POI/S, which signals a risk for subsequent POF, can be identified by measurement of anti-müllerian hormone (AMH) and follicle-stimulating hormone (FSH) (Rohr, 2008). POI/S in association with an intermediate size repeat has been reported (Streuli et al., 2009).5 Practically speaking, it might be advisable to have children earlier rather than later in life. It is not currently practicable to offer young women, as a routine, ovarian biopsy for cryopreservation of oöcytes for possible future use. Of course an ovum donated by a proven noncarrier sister would avoid the genetic risk. Female with Full Mutation. Offspring who inherit the fragile XA locus will all have full mutations. Hence, of their children with the FMR1 mutation, all the boys will have FXS, as will about 60% of the girls. The intellectual disability in these affected females is usually less severe than in the male.
A recent suggestion is that the heterozygous female child may run an additional risk, of ~5%, for X/XX mosaic Turner syndrome, possibly due to mitotic loss of the abnormal X chromosome in early embryonic development (Dobkin et al., 2009). Gray Zone Alleles. The possible pathogenicity of gray zones alleles has yet to be defined (see earlier). Expansion to a larger allele in a succeeding generation is rare, but recorded. F RAXE
The fragile XE story is incomplete, and referral to a specialist in this area is necessary. For the time being, counseling should include the following caveats: (1) the penetrance of intellectual disability in males and females is unclear, but it may be lower than for fragile XA syndrome; (2) the severity of intellectual disability is less in both sexes than for fragile XA; (3) variation between and within families appears to be considerable; and (4) a notable distinction from fragile XA is that daughters of fragile XE men can have full mutations. The phenotypic consequences of inheriting a full mutation are unclear. There is some risk of females being mildly handicapped, as yet unquantified, but probably less than for fragile XA. Some males with full mutations appear to be relatively normal, although not all have had detailed assessments. Nevertheless, we presume the risk of significant intellectual impairment in the male to be substantial, albeit less than 100%. Until the situation becomes clearer it may be best to offer prenatal diagnosis and present what is currently known to the couples involved. We await a more detailed interpretation of findings at prenatal diagnosis, but the picture is likely to be similar to FRAXA.
Prenatal Diagnosis F RAXA ( FMR1 )
Preimplantation Diagnosis. Given the high genetic risks that may apply to the female heterozygote, preimplantation genetic diagnosis (PGD)
5 An extraordinary circumstance is described in Rybak et al. (2009), concerning 32- and 26-year-old sisters, each with a 45-CCG repeat. The younger sister, having created embryos for her own future use, planned to undergo a second cycle in order to enable ovum donation for her older sister, who had already undergone menopause. The authors acknowledged an imperative “to chart a course balancing autonomy and altruism with the best interests of both recipient and donor and thereby attempt to navigate this dilemma in conformity with the ideal: first, do no harm.”
The Fragile X Syndromes •
253
would seem, in principle, an attractive proposition. From the single (or two) biopsied blastomere, PCR of the CCG repeat region, and of a number of polymorphic markers that can define the linked haplotype, is performed (Reches et al., 2009). However, it may be difficult to obtain adequate numbers of good ova from FRAXA carriers (note the earlier comments about primary ovarian insufficiency). The prognosis for success at PGD is related to repeat size: carriers with less than 100 CCG repeat copies suffer a more impaired ovarian response and decreased fertilization rate than do those with larger premutations and full mutations (Bibi et al., 2010). Ideally only embryos with normal alleles should be transferred, since the behavior of small premutations very early in development is unclear, and it cannot be excluded that expansions to a full mutation might occur later in embryonic development. Prenatal Diagnosis. Chorion villus sampling (CVS) is the preferred approach. Southern analysis requires a larger DNA sample and takes more time to complete than the many PCR-based molecular diagnoses carried out for a range of inherited disorders. Times conservatively quoted for a result from Southern analysis could be 2–4 weeks (compared with 2–3 days for PCR-based results). Sufficient DNA must be extracted from the CVS sample for at least one digest (approximately 10 μg), and with sufficient additional tissue to initiate a cell culture as a source of backup DNA. Diagnosis is based on repeat length; methylation status of CVS can be misleading, and nonmethylation of CVS has been associated with methylation in fetal tissues (Castellví-bel et al., 1995). Amniocentesis is not recommended: it is done at a later gestational stage and then takes further time for cell culture to provide enough DNA. The possible outcomes of prenatal diagnoses for fragile XA are as follows: (1) A normal male fetus (triplet-repeat copy number 6–54) (2) A male fetus with a premutation (approximately 55–200 copies of triplet-repeat), and thus a male carrier is predicted. There is a risk, the level of which can be estimated from Table 15–2, for FXTAS. (3) An affected male fetus with a nonmosaic full mutation, greater than 200 triplet-repeats. The FXS mental retardation syndrome is predicted. The phenotype of an affected male fetus with copy number mosaicism, that is, a mixture of full and premutations cannot be accurately predicted from 254
•
CVS; most cases would be affected, at least to some extent. (4) A normal female fetus (triplet-repeat copy number 6–54) (5) A female fetus with a premutation (approximately 55–200 triplet-repeat copies) and thus a female carrier is predicted. She would have a risk of premature ovarian failure and FXTAS. (6) A female fetus with a full mutation (greater than 200 triplet-repeat copies in most cells). Mental impairment, of variable degree, is predicted in at least half of full-mutation females.
F RA XE ( FMR2 )
There is little experience in the prenatal diagnosis of FRAXE, and expert advice should be sought. Treatment of Fragile XA Syndrome. The symptoms of FXS have been treated with a wide variety of behavioral and individual therapies, with varying success. Drug treatments have included stimulants and alpha-adrenergic receptor agonists, selective serotonin reuptake inhibitors (SSRIs), acetylcholine esterase inhibitors, antipsychotics for behavioral improvement, melatonin for disordered sleep, and anticonvulsants for seizures (see Chochaiya et al., 2009; D’Hulst and Kooy, 2009; Hagerman et al., 2009; and Kesler et al., 2009, for reviews). A specific rational treatment, aimed at the underlying cause of the disorder—in other words, attempting to arrest or reverse its aberrant pathophysiology— will require a good knowledge of how the lack of FMRP actually leads to neuronal dysfunction (Darnell et al., 2009). An example of this approach is to target one of the proteins normally subject to down-regulation by FMRP, and the metabotropic glutamate receptor 5 (mGluR5) is a case in point. In the absence of FMRP, an excessive amount of mGluR5 is produced, and this leads to a “long-term depression” of synaptic connectivity. Fenobam is an mGluR5-antagonist, and a trial of this drug in FXS patients is underway (Berry-Kravis et al., 2009). Another drug that may be trialed is minocycline, a tetracycline analog, which influences dendritic spine maturation in an animal model (Bilousova et al., 2009). In spite of some promise for future treatment of FXS individuals, it is improbable there will ever be a cure, but nevertheless these treatments may well have a helpful role to play in the lives of parents, carers, and the FXS individuals themselves.
PA R E N T W I T H A C H R O M O S O M A L A B N O R M A L I T Y
PART THREE VARIANTS
This page intentionally left blank
16 variant chromosomes and abnormalities of no phenotypic consequence
IN THIS CHAPTER we deal with three kinds of classical chromosomal variation, having the quality in common that typically they carry no implications for abnormality in the person. First, there is the matter of normal variation. Normal chromosomes do not necessarily look exactly alike in different individuals, and some chromosomes show a remarkable degree of variation in their morphology. Obviously enough, it is crucial that the cytogeneticist distinguishes normal variation from abnormality. Generally, there is no point in reporting a particular variant to the referring practitioner or to the patient. But it is sometimes necessary to pursue the matter, with family studies, when it is not clear whether a particular finding is a normal variant or an abnormality, or when a previous report has sown a seed of doubt in the patient’s mind. The study of normal variants is also a research activity in its own right. Second, we bring together in this chapter a group somewhat flowing on from the first, comprising
structural rearrangements due to the translocation of harmless material, such as heterochromatin and nucleolar organizing regions, from one chromosome to another. Third, we treat a group categorized as the “euchromatic abnormality without phenotypic effect.” These are chromosomal differences that do not too comfortably accommodate the expressions “normal” and “variant,” and yet “abnormality” may convey too strong a sense. Anomaly may be a better word, or if “abnormality” is to be used, it should retain, in the reader’s mind (if not in the text hereafter), its quotation marks. These are deletions and duplications that might at first sight have been thought to be abnormalities that would be associated with some clinical defect—deletions up to 16 Mb and duplications as large as 30 Mb in size—but which are in fact observed in normal persons. Finally, in the new era of microarrays, copy number variants loom large as a new form of variation, and this demands its own chapter (which follows this one).
•
257
BIOLOGY CHROMOSOME VARIANTS If, in an individual, one chromosome of a homologous pair looks normal and the other has a different cytogenetic appearance, the pair may be said to be heteromorphic. A collection of such heteromorphisms detected with a variety of techniques is provided in Table 1 of ISCN 2009. Cytogenetic differences across the whole pool of chromosomes are referred to as variants.1 Variants may be frequent or rare. The classical chromosome variants present a continuous spectrum, rather than a bimodal distribution. For example, considering the differing lengths of the short arms of the acrocentric chromosomes, these could be arbitrarily classified as being of approximately typical length, somewhat shorter, somewhat longer, very short, or a lot longer. They are sufficiently variable that they were exploited as markers in research (in the days before DNA markers had been well developed). Some are so rare they qualify as private variants, recorded in just a single family. We consider chromosome variation in these three areas: the centromeric heterochromatin, the acrocentric short arms, and the fragile sites. C-BAND SIZE, POSITION, AND STAI N IN G P R O P ERT I ES
C-band heterochromatin comprises, by definition, permanently inactive DNA (constitutive heterochromatin), and it is mostly located adjacent to the centromere (C for constitutive). It stains darkly on C-banding. The four originally described variant forms are 1qh, 9qh, 16qh, and Yqh (qh for long arm heterochromatin); the differences in size were great enough to have been detected on solid-stain chromosomes in the prebanding era. C-bands vary in size, and, for those chromosomes in which the material is centromerically placed, there is variation in position relative to the centromere (Craig-Holmes, 1977). The position of the centromere within the C-band positive heterochromatin block of the 1qh,
9qh, and 16qh may vary due to inversions of the heterochromatin. The observed frequencies vary according to the precision of staining and the criteria of the observer (Kaiser, 1988). The commonest of these variant inversions— and indeed the commonest chromosomal variant in the human race—is the placement of 9q heterochromatin into 9p, immediately adjacent to the centromere. This high frequency presumably reflects the existence within the pericentromeric region of a number of hotspots for recombination (Starke et al., 2002). In about half of no. 9 chromosomes, a small amount (less than one-third) of the heterochromatic block is sited in the short arm. In about 10%, there is a “partial” inversion, with about one-third of the heterochromatin in the short arm, inv(9) (p11q12). In 0.6%, all the heterochromatin is in the short arm—a total inversion, inv(9)(p11q13). The wide possible range is presented in Starke et al. (2002). Possibly the most impressive 9qh variant is that recorded in Lukusa et al. (2000), in which the segment 9p11-q13 had undergone a tandem duplication. The abnormal chromosome at first sight might have offered a diagnosis for the mental defect of the woman in whom it was discovered, but her normal sister had the same chromosome, effectively exonerating it. Whether an “inversion variant” chromosome can influence its own disjunction is speculative. Willatt et al. (1992) observe that in 11 reported cases of the rare mosaic trisomy 9 syndrome, four occurred in the setting of maternal heterozygosity for the inv(9)(p11q12) variant and suggest a causal link; and partial trisomy 9 has been associated with a parental inv(9) (p11q12/3) (Kaiser, 1984; Stamberg and Thomas, 1986). These tiny numbers need to be seen against the background of the hundreds of thousands of inv(9) heterozygotes who have not come to cytogenetic attention for such a reason. Fortuitous coincidence, or ascertainment bias, remain a perfectly acceptable explanation.
1 The word polymorphism is also used as a cytogenetic colloquialism in this context, but, to be precise, it is an inaccurate usage. The genetic term polymorphism refers to the presence of an uncommon allele that has a frequency of >1% in the population, and although some chromosome variants may be more common, each one (e.g., 9qh+) is not necessarily homogeneous but may comprise a collection of many “alleles” at this locus. Darwin had this comment, from his Origin of Species: “Variations neither useful nor injurious would not be affected by natural selection, and would be left a fluctuating element, as perhaps we see in the species called polymorphic.”
258
•
VA R I A N T S
Variation of C-band material can be demonstrated using other staining techniques, such as Q-banding, which reveal differing intensity of fluorescence. Most notably, this variation is seen in the C-bands of chromosomes 3 and 4, which range from very dull to very bright. The staining of the C-band material, after one round of replication in BrdU, varies in the pattern of lateral symmetry (Angell and Jacobs, 1978). Heteromorphic staining of the centromeric regions of all chromosomes except chromosome 8 has been demonstrated using various restriction endonucleases to treat the chromosomes prior to Giemsa staining (Babu et al., 1988). Jabs and Carpenter (1988) showed that the extra C-band material adjacent to the centromere on the short arm of a variant chromosome 6 is due to increased amounts of a chromosome-specific alphoid DNA repeat sequence. Other unusual C-band variants are reported for chromosome 3 (Petrovic, 1988), chromosome 5 (Fineman et al., 1989), chromosome 11 (Aiello et al., 1994), chromosome 12 (De Pater et al., 2006), chromosome 18 (Pittalis et al., 1994), chromosome 20 with increased C-band material in the short arm (Fryns et al., 1988), and chromosome 20 long arm (Romain et al., 1991). The 20q variation reflects different amounts of chromosome-specific alphoid DNA. A duplication of the centromere itself is a different entity (Callen et al., 1990; Till et al., 1991). Morphological variants of the Y chromosome are in two categories. (1) Continuous variation in the amount of C-band-positive heterochromatin can range from a virtual absence, in which case the Y chromosome may appear to be only about half the size of chromosome 22, to a large amount such that the chromosome is about the same size as chromosome 13. Paternal chromosome study is worthwhile to confirm that very small chromosomes are in fact variants (short Y chromosomes with breakpoints proximal to the Yq11/12 heterochromatin interface are pathogenic deletions; Salo et al., 1995). For very large ones, C-banding is adequate to confirm that the increased size is due to heterochromatin. (2) Discontinuous variation in the Y chromosome is expressed as a metacentric appearance, presumably due to pericentric inversion. Satellites on the end of the long arm are another variant; these are presumably due to translocation from one of the acrocentric chromosomes and have been documented to segregate in large kindreds, over centuries (Genest, 1973) (and see later in the section “NOR Translocation”).
These discontinuous variants are normally without phenotypic effect, but, again, paternal chromosome study is warranted if there is any doubt. Clinical Significance. Many studies have purported to show that variant chromosomes involving C-band size and position are associated with congenital malformations, cancer, recurrent pregnancy loss, and infertility. There have equally been many studies that report no such association. Carothers et al. (1982) concluded, and we concur, that “reproductive fitness of carriers of heterochromatic variants of the human karyotype is normal.” ACROCENTRIC SHORT ARM M O R PH O LO GY
The short arms of the acrocentric chromosomes show a range of morphology, reflecting variation in three components of the short arm: the centromeric heterochromatin, the satellite stalk, and the satellite material (Fig. 16–1). These three components correlate with the three bands p11, p12, and p13. The p11 band contains several types of tandem DNA repeats, including satellites I, II, II and beta satellite; band p12 contains multiple copies of genes coding for ribosomal RNA (rRNA); and band p13 incorporates beta satellite (Piccini et al., 2001;
Satellite Satellite stalk Proximal short arm Centromere
Long arm
FIGURE 16–1 Diagram of an acrocentric chromosome showing the variable components: satellite, satellite stalk, proximal short arm, and centromere.
Abnormalities of No Phenotypic Consequence •
259
Bandyopadhyay et al., 2001b). The nucleolus of the cell is formed by an aggregation of rRNA; thus, the stalk (band p12) is sometimes called the nucleolar organizing region (NOR). This stalk stains darkly with silver nitrate (Ag-NOR staining) on those acrocentric chromosomes that have an active NOR; and the picture varies from no uptake through marked uptake (darkness) of stain. A 3.3 kb repeat sequence, having homology to the repetitive DNA D4Z4 located at distal 4q, is found on the short arms of all the acrocentric chromosomes; and euchromatic-like sequences and transcripts exist in the short arm of chromosome 21 (Lyle et al., 2007). Length and satellite size and number are distinguishing features. At one extreme, a short arm may seem to be absent; at the other extreme, it may be so long that a D-group chromosome is of C-group appearance, and a G-group chromosome has an F-group resemblance. Possibly the largest acrocentric short arm ever seen is the chromosome 15 described in Friedrich et al. (1996) in which the “short” arm was actually longer than the long arm. Molecular analysis of one chromosome 14 with an apparently absent short arm showed loss of satellite III DNA (Earle et al., 1992). Satellites vary widely in appearance: apparently absent, small or large, and single or double. Variation can occur within the one individual (Ozen et al., 1995). A meiotic exchange can lead to a parent’s variant satellite appearing on another chromosome in the child (Farrell et al., 1993)—the “jumping satellites of Gimelli.” The short arm of chromosome 15 stains brightly with DA/DAPI, but 18% of individuals in an English patient population, with a range of referral reasons, had extra DA-DAPI signals on other acrocentric chromosomes, 60% of them on chromosome 14; thus, an acrocentric chromosome may have its short arm originating from another acrocentric (Cockwell et al., 2007). An apparent large acrocentric short arm can rarely mislead. Benzacken et al. (2001b) report a ‘14p+’ chromosome discovered at late pregnancy amniocentesis (36 weeks), a fetal macrosomy having been seen on ultrasonography, and the 14p+ initially considered to be a normal variant. When the child was born, a diagnosis of Beckwith-Wiedemann syndrome (BWS) was confirmed clinically, and the infant’s karyotype was reassessed. The appearance of the NORs was judged, on this occasion, to be not quite typical. Fluorescence in situ hybridization (FISH) with 11p probes convincingly showed a rcp(11;14)(p15;p13), demonstrating that the 260
•
VA R I A N T S
abnormal chromosome was a der(14), and that the BWS was due to a partial 11p trisomy. The parents’ chromosomes were normal. De Pater et al. (2000) describe a similar circumstance, in which a prenatally diagnosed de novo 14p+ “polymorphism,” which was negative on NOR staining, turned out actually to be a t(14;17)(p11;p11), and the child, who died as a neonate, thus had 17p trisomy. Clinical Significance. Hassold et al. (1987) could demonstrate no association of NOR variants with an increased risk for trisomic conception, a possibility that had previously been postulated. An increased risk to have a child with Down syndrome had previously been proposed for the person with a “double NOR” on an acrocentric chromosome. This was a reasonable postulate, given the in vitro cytogenetic observation of “satellite association” and imagining that a similar phenomenon could happen in vivo in meiosis, predisposing to aberrant segregation, and that this would be more likely with larger satellites. But the actual observation is, in fact, of no such increased risk (Serra and Bova, 1990). The possibility that translocated acrocentric short arms or compound centromeres might predispose to nondisjunction and miscarriage (Cockwell et al., 2003) has not been borne out by more extensive investigations ( J .C. K. Barber, personal communication, 2010) F RA G I L E S I T ES
A fragile site is a point on a chromosome that is liable, upon classical cytogenetics, to show gaps and breaks. The location of the fragile site is the same in all cells in a particular individual or family (Sutherland and Hecht, 1985). Fragile sites are classified on the basis of their frequency in the population and the conditions of tissue culture that are required to induce them. In terms of frequency (ignoring fragile XA and fragile XE) there are three categories of fragile site (Table 16–1). (1) Rare sites are present only in one per several hundred individuals. (2) Two fragile sites are of intermediate frequency: fra(10q25), seen in 2.5% of individuals, and fra(16q22), seen in 1%–5% of individuals. (3) So-called common fragile sites are universal and form part of normal chromosomal architecture, and in which the underlying cause is hypoacetylation of histones in this region of the chromosome ( Jiang et al., 2009). Only the rare and intermediate fragile sites are classified as
Table 16–1. The Three Groups of Rare Variant Autosomal Fragile Sites Folate-sensitive 1p21.3 2p11.2 2q11.2 2q13 2q22.3 5q35 6p23
7p11.2 8q22.3 9p21.1 9q32 10q23.3 11q13.3 22q13
BrdU-inducible 12q24.2 10q25a Distamycin A-inducible 16p12.1b 8q24.1b b 16q22.1a 11p15.1 Unclassified 15q13c
11q23.3 12q13.1 12q24.13 16p13.11 19p13 20p11.23
17p12
a
Of intermediate frequency. Recorded in Japanese populations only. c Recorded in the Turkish population only Source: From Sutherland (2003). b
chromosome variants (Fig. 16–2). The common fragile sites, being universal, by definition do not vary; although the proportion of metaphases in which they are seen can vary, depending upon conditions of cell culture, from 0 to 20%. FRA11B is the fragile site at 11q23.3, within the proto-oncogene CBL2, and which displays a molecular behavior similar to FRAXA ( Jones et al., 1994). Jacobsen syndrome comprises variable deletions of 11q, and it may be that mothers of some affected children have a cytosine-cytosine-guanine (CCG) triplet expansion at FRA11B. However, in the majority of these children the deletion breakpoint is at a site elsewhere than at 11q23.3 (Penny et al., 1995; Michaelis et al., 1998). Clinical Significance. Fragile sites are harmless (except, of course, fragile XA and fragile XE, and, remotely possibly, FRA11B). The fragile sites of rare and intermediate frequency have been described variously as being associated with congenital malformations, sporadic chromosome abnormalities, and a predisposition to cancer. But, as noted earlier, there is no convincing evidence of any of these associations existing other than by chance (Sutherland and Baker, 2000). There is no apparent relation
between autosomal fragile sites and mental retardation (Fryns and Petit, 1987). The CCG repeat at 11q23.3 might conceivably be a vulnerable point allowing de novo deletion of 11q ( Jacobsen syndrome), but the evidence for this is by no means compelling. The fra(10)(q23) may lead to a del(10) (q23) being detected at prenatal diagnosis; this observation appears to be without phenotypic consequence (Zaslav et al., 2002). There is the clinical interest that one form of Seckel syndrome (p. 347) displays chromosomal breakage at fragile sites (Casper et al., 2004). Fragile sites that have been characterized at the DNA level are all in genes that are silenced by hypermethylation when the fragile site is present. Theoretically, homozygosity might have been expected to result in an abnormal phenotype, depending upon the nature of the gene involved; but in fact no such homozygosity has been recorded. Homozygosity for some folate-insensitive fragile sites (10q25, 16q22, 17p12) is described; the individuals studied had no consistent phenotypic abnormality, possibly because these fragile sites are located in highly repetitive DNA areas devoid of genes. Y HET ER O CH R O M AT I N T R A N S LO CAT I O N S
In the sense that they cause no phenotypic abnormality, the so-called t(Y;15) and t(Y;22) translocations, in which the C-band material from the long arm of the Y has been translocated onto the short arm of the acrocentric, can be regarded as examples of normal heteromorphism (Friedrich and Nielsen, 1972; Cohen et al., 1981; Neumann et al., 1992). Once formed, these variant chromosomes are stable. In one reported family, a girl with Prader-Willi syndrome had a de novo 15q11-q13 microdeletion on a familial t(Y;15)(q12;p11) chromosome. Her father, brother, uncle, and two aunts also carried the intact t(Y;15) (Eliez et al., 1997). Ascertainment bias is the more likely explanation of such coincidences. A segment of Y chromosome heterochromatin can be insertionally translocated into another chromosome, and this may be without any phenotypic effect. Ashton-Prolla et al. (1997) report detecting such an anomaly at prenatal diagnosis, with the karyotype only interpretable after the father had been studied. He had a segment from Yq12 inserted into chromosome 11 at band 11q24: 46,XY,der(11) ins(11;Y)(q24;q12q12). He had inherited this ins(11;Y) from his mother, and her normality, and subsequently his infant daughter’s normality, attested Abnormalities of No Phenotypic Consequence •
261
FIGURE 16–2 Variant autosomal fragile sites and the fragile X (FRAXA). The chromosome on the left of each pair is plain stained, and the one on the right is G banded.
to the innocuousness of this variant chromosome. A similar case is described in Spak et al. (1989), of a harmless C-band positive insertion at 11q23.2; the origin of the heterochromatin could have been an autosome or the Y chromosome. Clinical Significance. These Y heterochromatin translocations are to be regarded as variants without phenotypic consequence. NOR TR AN S LO C ATION
The NOR can be translocated to another chromosome, usually to a terminal region; the nomenclature of ps or qs denotes a satellite (that is, a NOR) at 262
•
VA R I A N T S
the tip of the arm in question. Willatt et al. (2001a) document examples of this phenomenon, either as terminal or as interstitial NORs, in chromosomes 1, 2, 4 (several cases), 6, 7, 8, 10, 12, 17, and 22, and Faivre et al. (1999) discuss the recorded cases identified at prenatal diagnosis, noting that in only one of 13 was there an otherwise unexplained abnormal phenotype, and acknowledging that this could have been a coincidental association. The anomalous chromosome can arise somatically, as Storto et al. (1999) show in a father with 46,XY/46,XY,10qs mosaicism, whose (normal) child had been diagnosed with the 10qs at amniocentesis. Wilkinson and Crolla (1993) showed, in the Yqs chromosomes segregating in three families, that the
NOR on the Y in each had arisen from a 15p to Yq translocation, as did two of the four cases of Kühl et al. (2001). These Yqs chromosomes have lost their pseudoautosomal region-2, but this is of no discernible clinical consequence. Reddy and Sulcova (1998b) undertook detailed molecular dissections of three NOR translocations: a chromosome 21 into the short arm of which segments of chromosome 15 beta-satellite elements were inserted, making it “tricentric”; a chromosome 7 with acrocentric betasatellite DNA inserted at 7p13; and a pseudodicentric chromosome 2 with beta-satellite inserted at the tip of 2q. Notwithstanding the typical circumstance of harmlessness, the NOR translocation may, in rare instances, and if there is a deletion or disruption at the point of attachment, be pathogenic. Chen et al. (2000) report their experience with a “4ps” in a mother and Wolf-Hirschhorn syndrome in her child, but in fact this reflected unbalanced segregation from a reciprocal translocation t(4;15)(p16;p11.1). Their second case of an Xqs chromosome is more telling, in which a 46,X,Xqs mother had an abnormal 46,Xqs,Y son. They proved the point that there was a distal Xq deletion, and so the abnormal chromosome might more accurately be described as Xpter→Xq28::sat, rather than Xpter→Xqter::sat. A notable example of a possibly pathogenic insertional NOR translocation is that described in Tamagaki et al. (2000) (and see p. 189), in which an NOR inserted interstitially at Xq11.2 was segregating with a spastic paraplegia in the males. The cosegregation could be coincidental, but equally might reflect disruption of a causative X-borne locus due to the NOR insertion. Clinical Significance. In general, these satellited chromosomes are to be regarded as harmless heteromorphisms. Faivre et al. (2000a) reasonably comment, in their study of a familial 4qs ascertained through a child with a cerebellar ataxia, that “genetic counseling should be reassuring” in the setting of an NOR translocation identified at prenatal diagnosis. We note earlier rare exceptions to this rule, in which the translocated NOR may have caused genomic disruption at the site of translocation. The lesson from these cases is that any satellited chromosome deserves detailed study (especially an X chromosome NOR not known to be carried by a phenotypically normal male in the family), while retaining the perspective that, very probably, the conclusion will be that it truly is a harmless variant. Subtelomere
FISH or microarray analysis may be reassuring to a family with a de novo telomere-NOR translocation, by demonstrating that no material of consequence is missing.
CHROMOSOME ANOMALIES Euchromatic Abnormalities of No, or Uncertain, Phenotypic Consequence The G-band pattern is generally constant, and the relative sizes of G-bands are similar between the karyotypes of any members of the human race. The fact of being a diploid species is useful, enabling one homolog to be compared with the other in any metaphase. Most of the banding pattern variation observed by the cytogeneticist is artifactual. The level of resolution, which is a function of the degree of compaction of the chromatin, will determine the number of bands seen in any metaphase, and even within a metaphase the number of bands on homologous chromosomes may vary (“homolog asynchrony”). Two major categories, at the cytogenetic level, may be considered (Barber, 2005) (the category due to molecular-level copy number variation is treated in the next chapter): • Euchromatic variants due to constitutional cytogenetic amplification • Euchromatic deletions and duplications with no, or uncertain, phenotypic effect A distinction between one of these anomalies and a pathogenic cytogenetic abnormality may not always be easy. It is generally considered that if, in a family study, all the individuals (with the usually allowable exception of the index case) having the unusual chromosome are phenotypically normal, then the chromosome really is a euchromatic abnormality of no phenotypic consequence. And yet, some chromosome anomalies, judged to be harmless by this criterion, might have a euchromatic deletion or duplication that is actually of similar extent to some of those which are undoubtedly pathogenic. How can there be such a difference in outcome: a normal or an abnormal individual? What matters is the nature of the loci in the segment of interest. Is this a region of low gene content? Harmless deletions frequently involve G-band dark euchromatin with relatively low gene density (Barber et al., 2005; Daniel et al., 2007), but at least one harmless duplication of a region of moderate gene density has been Abnormalities of No Phenotypic Consequence •
263
described (Barber et al., 2007). Are the genes dosage sensitive? If a gene, or a set of genes, function equally well at a 50% as at a 100% level, a deletion on one chromosome will be without any effect upon phenotypic expression and could be thought of as being “haplo sufficient.” Are the genes part of a family of genes or a network of genes of related function? If so, it is possible that other genes in the same family or network may compensate for the loss of one of their members (Barber et al., 2005, 2006). Rarely, a parent-of-origin effect is invoked (Temple et al., 1996). Finally, the interpretation of phenotypic normality needs to made with some confidence. It could be that a deletion or duplication in a gene-sparse region might lead to only a very subtle compromise of form and function, just within the range of normality; but more detailed clinical assessment could show that the chromosome abnormality is actually, if subtly, pathogenic (Lopez-Exposito et al., 2008). E U CHRO MATIC VARI A N TS
In seven chromosome regions, an additional band or bands may infrequently be discerned. These bands reflect changes of known copy number variable regions, as listed in the Database of Genome Variants (http://projects.tcag.ca/variation). When the copy number is high enough, they become visible in the light microscope and may be described as “euchromatic variants”; Barber (2008a) also refers to “chromosomal copy number variants.” An additional large tract of segmentally duplicated euchromatin may have a similar appearance (Willatt et al., 2007). The regions of interest comprise 4p16, 8p23.1, 9p12, 9q12, 9q13, 15q11.2, and 16p11.2. With few exceptions, no major clinical significance attaches to the discovery of any of these variants (Barber, 2005). We provide brief commentaries on each. Band 4p16.1. A cytogenetically visible amplification of 4p16.1 cosegregated with ear and eye defects in a large family reported by Balikova et al. (2008). Five tandem copies of a 750 kb amplicon were present on the variant chromosome. The amplicon contained olfactory receptor, defensin, and DUB gene family members, which are known, individually, to be copy number variable. The amplicon in this family may have been the basis of the defects inherited in a mendelian fashion (in which case, euchromatic “variant” would be a misnomer) but, alternatively, the variant may simply have been cosegregating with a closely linked mutation. 264
•
VA R I A N T S
Band 8p23.1. A number of families with euchromatic variants of 8p23.1 and a normal phenotype have been described (Barber et al., 1998, Barber, 2005). The anomalous G-light band appears larger with a fine G-dark band in the middle of the enlarged band. The additional material contains multiple copies of a 260 kb amplicon of olfactory receptor and defensin genes (Hollox et al., 2003). Variant carriers have a total of 8–12 copies of this amplicon compared with the 2–7 copies found in the general population. Copy number variation of this region is normally benign, except for a predisposition to psoriasis with increasing copy number (Hollox et al., 2008). Predisposition to Crohn’s disease from reduced (Fellermann et al., 2006) or increased copy number (Bentley et al., 2010) has not been confirmed in larger series of patients (Aldhous et al., 2010). Distinction is to be made from the known pathogenic 8p23.1 duplication, which is cytogenetically indistinguishable from the euchromatic variant and which can arise de novo, or be transmitted from parent to child (Barber et al., 2008; Barber et al., 2010). Pericentromeric Chromosome 9. There are at least four kinds of euchromatic variants of chromosome 9 which reflect the existence of pericentromeric segmental duplications that are themselves copy-number variable (Redon et al., 2006). In the short arm, an extra euchromatic band is seen between 9p11 and 9p13. This may be due either to multiple copies of a ~1 Mb amplicon (Lecce et al., 2006) or to extra copies of large tracts of segmentally duplicated material (Willatt et al., 2007). In the long arm, extra bands may exist within the major block of long arm heterochromatin in 9q12/9qh, or in the proximal long arm between 9q13 and 9q21.2; deletion of 9q13 to 9q21.12 (and perhaps 9q21.2) may also be observed ( Joseph-George et al., 2011). Duplication, triplication, or deletion of segmentally duplicated material accounts for the extra or missing bands. No clear phenotypic effects have been demonstrated and the suggestion of an association with recurrent pregnancy loss remains seems unlikely (Dundar et al., 2008; Barber, 2010). Band 15q11.2. The importance of these 15q11.2 euchromatic variants lies in the need to distinguish them from the pathogenic 15q11.2–15q13 duplications (and triplications) of the Prader-Willi/ Angelman critical region (PWACR) (p. 329), which can look very similar under the light microscope
(Barber, 2005). The extra band (or bands) reflects amplification of a cassette of pseudogenes derived from the neurofibromatosis type 1 (NF1), immunoglobulin heavy chain D (IgH D), gamma-amino-butyric acid receptor 5 (GABRA5), and B-cell lymphoma genes (BCL8) (Fantes et al., 2002). These are located between the centromere and the most proximal breakpoint (BP1) of the PWACR. Copy number of the NF1 and GABRA5 pseudogenes is 1–4 in normal controls, and 5–10 in variants. Band 16p11.2. A number of families are known with an apparent duplication of band 16p11.2 (Barber, 2005; Lopez Pajares et al., 2006). The euchromatic variant, which lies practically adjacent to the pericentromeric heterochromatin, can be extensive, almost doubling the overall size of the short arm. The extra band is due to amplification of a pseudogene cassette that is derived from immunoglobulin heavy chain D (IgH D), creatine transporter (SLC6A8), and adrenoleukodystrophy (ALD) genes. Copy number in controls has been estimated as 4 and between 10 and 12 in variant carriers. Distinction is to be made from pathogenic duplications of 16p11.2 (p. 330). Band 16q11.2. An additional G-light band within the major 16q11.2/16qh block of heterochromatin may result from an inverted duplication of the centromere ( Jalal et al., 1990). This needs to distinguished from pathogenic duplications of 16q12.1-q13 (Barber et al., 2006), which can appear to be inserted into the 16q11.2/16qh block. E U CHRO MATIC D UP LI CAT I ONS A N D D E L E T IO N S W ITH N O, OR U NCE RTA I N , P HE NOTY P E EFF EC T
Thinking purely in mechanical and structural terms, these duplications and deletions of little or no phenotypic effect may be no different, in terms of structure, from a pathogenic abnormality of similar size. That is, a similar process has led to the addition or removal of a segment of chromatin. As discussed earlier, it is likely that the nature of the segment in question is what determines harmlessness or pathogenicity: put simply, a gain or a loss of genes that does or does not matter. Or, if it does matter, it may matter very little, and the effect of the gain or loss may be so mild that the phenotype is only subtly influenced, which thus remains well within the range of normality.
Quite a number of these cytogenetically visible duplications and deletions have been reported (and several more unpublished examples are likely known in a number of cytogenetics laboratories). Barber (2005) has undertaken a review, and Table 16–2 sets out the harmless deletions and duplications that have been recorded. Most of these unusual chromosomes had been detected in normal persons, and the same then in normal relatives; prenatal diagnosis is also a frequent route of ascertainment. In other cases (Table 16–3), the chromosome was first discovered in an abnormal individual, but then the same chromosome was identified in other phenotypically normal relatives. This observation of normality in other family members would typically allow the presumption of the chromosome being simply a variant, although one should be careful about extrapolation to different families in which the same unbalanced chromosome abnormality is observed. In a few families, the child’s abnormality may have been the direct result of the “chromosomal variant,” the parent’s normality notwithstanding, and a number of explanations can be proposed. Variable penetrance and expressivity, more traditionally invoked in mendelian genetics, are gaining a broader acceptance in cytogenetics, with the 1q21.1 microdeletion in thrombocytopenia absent radius (TAR) syndrome a notable example; Klopocki et al. (2007) propose an unidentified “modifier” of expression to explain their finding that, in 75% of families, the clinically normal parents will have the same 1q21.1 microdeletion (see also p. 311). A similar explanation may apply to distal 3p deletions that may or may not lead to a phenotype (Barber, 2008b). The distinction between a euchromatic abnormality of no phenotypic consequence and one of only slight consequence may be a very subtle one. Consider the case of a girl described in Bonaglia et al. (2002) who had a karyotype ordered on fairly slender grounds at age 18 months (she was in hospital at the time for an ear infection), but as a 6½ year old had only the most minor signs (one ear ½ cm longer than the other, and crowded teeth, for example), and IQ testing at 5½ years had shown an average intelligence. She had a de novo dup(9) (p22→p13), proven to have arisen following an asymmetric sister chromatid exchange at paternal meiosis. Would she have been slightly different in appearance, her dentition less imperfect, and her IQ a little higher, if she had had an apparently normal karyotype 46,XX? Or would she have been just the same, and the dup(9p) was truly a euchromatic Abnormalities of No Phenotypic Consequence •
265
Table 16–2. Euchromatic Duplications, Deletions, and “Variants” without Phenotypic Effect, as Inferred from the Observation of Transmission from Phenotypically Normal Parent to Normal Child CHROMOSOME
del
1 2 3 4 5 6 7 8 9
10 11 12 13
dup
VARIANT
p21-p31 q31.1-q32 p12-p12 q13-q14.1 p25.3-pter q34.1-q34.3 p14-p14 p25-pter q22.31 - q23.1 p22.3-pter p23.1/2-pter q24.13-q24.22 p21.2-p22.1
q11.2q21.2 p12-p12 q14.3-q21.33
15 16 18
q13q22 p11.31-pter
22
p11.21-pter
q28-q29 p16.1p16.1
p23.1-p23.3
p23.1
p11.2-p13.1
p12 q12 q13-q21.2
p13-p14 q14.1q23.1 q21.31q22 q13-q14.3 q14-q21 q11.2 p11.2 p11.2-pter q11.2q12.2
Note: The estimated sizes of the deletions and duplications range from 4.2–16.0 Mb (del) and 3.4–31.3 Mb (dup). Source: From Barber (2005) with additional material from the Chromosome Anomaly Collection Web site. Updated information is posted in the “What’s New” section at http://www. ngrl.org.uk/Wessex/collection/index.htm
abnormality of no phenotypic consequence? Since dental crowding is seen in the classical 9p duplication syndrome, perhaps this aspect truly is due to the karyotypic imbalance. To recapitulate: at the fundamental level of cytogenetic mechanism, at least some of the abnormalities with and some without phenotypic effect may have arisen in essentially the same way, and the difference depends upon whether the genes duplicated or deleted were dosage sensitive (and recognition
266
•
VA R I A N T S
naturally depends upon whether their effects are observable at clinical examination). SU BT ELO M ER I C D EL ET I O N S A N D DU PL I CAT I O N S
Some “deletions” and “duplications” identified on subtelomeric FISH probing may well be true findings, but being without effect upon the phenotype (Riegel et al., 2001). Ballif et al. (2000a) record, for example, the following (sub)telomeric variants: ish
Table 16–3. Euchromatic Deletions or Duplications in Which the Child Was Abnormal But the Parent Normal CHROMOSOME del
dup
1
q11-q22 q42.11-q42.12
2 3
q13-q13 q14.1-q14.2 p25.3-pter
4 5 6 7 8 10 11 13 14 15 16 18 21
p15.3-pter p14.1-p14.3
p11.2-p12.3 q25-q25 q31.3-q33 q35.1-q35.2 q15-q21 q23.3-q24.2*
p22-pter p23.1-p23.2 q11.2q11.23 q14.3-q22.1 q14-q14**
p23.2-p23.2
q21.1-q21.32 q24.3-q31 q11-q13 q11-q12 q12.1-q12.1 q21.31-q22.2
q21.1-pter
Notes: The estimated sizes range from 3.6–10.0 Mb (del) and 2.0–16.3 Mb (dup). In several of these cases, the abnormality in the child is likely to have been coincidental, and the chromosomal abnormality was, of itself, without phenotypic effect. In some, however, a causal link must be presumed, such as the footnoted dup 6q24 and del 13q14, albeit in these two requiring the agency of another factor (imprinting effect; mutation on other homolog). *Child with transient neonatal diabetes, presumed to have been a consequence of paternal disomy for this segment (Temple et al., 1996). **Child had retinoblastoma, presumably due to a second hit at the RB locus at 13q14 on the other chromosome 13 in a retinal cell (Cowell et al., 1988). Source: From Barber (2005), with additional material from the Chromosome Anomaly Collection Web site. Updated information is accessible at http://www.ngrl.org. uk/Wessex/collection/index.htm
add(1)(13qtel+) using PAC probe 163C9; ish del(2)(qter) with PAC 1011O17 and P1 210E14; ish del (9)(pter) with PAC 43N6; and ish del(X) (pter) with cosmid CY29. The harmlessness (in all probability) of these variants was demonstrated, as per usual, by showing the same thing in a parent.
Many more such variants have been discovered, as subtelomeric analysis has become routine using FISH and/or multiplex ligation-dependent amplification (MLPA), and more surely will be, as the use of microarray-CGH becomes more widespread (Adeyinka et al., 2005; Ravnan et al., 2006; Balikova et al., 2007; Martin et al., 2007). The normal phenotype in many of these euchromatic subtelomeric deletions and duplications may possibly be explained by the degree to which they are composed of tandem duplications (Linardopoulou et al., 2005). Informed interpretation of these findings will require of the molecular cytogeneticist a ready ability to access information in the literature and on genome browsers, and an awareness of the best probes to use in order to distinguish between variants and true pathogenic abnormalities. Details of Meiotic Behavior. Variant chromosomes, being normal chromosomes, behave normally at meiosis; and other things being equal, 1:1 segregation occurs. Hence, any individual with a variant chromosome transmits it to, on average, half of his or her offspring. In the specific case of the inv(9)(p11q13), an essentially 1:1 ratio was confirmed in a sperm study (Colls et al., 1997). Similarly, most simple duplications and deletions are directly transmitted, intact, in a 1:1 ratio. Very rarely, an apparently nonpathogenic duplication in a parent can undergo replication that leads to a pathogenic triplication in a child, and Yobb et al. (2005) and López-Expósito et al. (2008) provide examples with respect to parental duplications of a 3 Mb segment in 22q11.2, and a 12 Mb segment in 13q21.1-q21.33, respectively.
GENETIC COUNSELING Reproductive Advice A person carrying a variant chromosome has, practically by definition, no increased risk for having abnormal offspring, pregnancy loss, or any other reproductive problem. Some see it as at best pointless and at worst counterproductive even to mention to the individual that a variant chromosome has been found; others feel obliged to pass on the observation. If it is discussed, it must be made clear that it is a normal finding: perhaps interesting, but of no practical importance. (Some patients may be intrigued to learn that they are of interest to genetic researchers.) For the size variants (C-band and Abnormalities of No Phenotypic Consequence •
267
NOR), the point can simply be made that some chromosomes come in short, medium, and long forms, and where a chromosome happens to fit in this continuum is without significance. There is considerable potential for iatrogenic anxiety, whereas in reality the biology of the supposed anomaly has no pathogenic implication. The counselor may thoroughly understand the presumed harmlessness of a variant chromosome, but the person in whose family it has been discovered may react “nonscientifically.” To put a stark setting, the worst possible response might be for a couple to choose to terminate a pregnancy because of an overinterpreted variant chromosome, as has actually happened with the 16p11.2 euchromatic variant (López Pajares et al., 2006). “Primum non nocere”: first do no harm. PARE N T W ITH SAME VA RI A NT A S ABNO R MA L C H ILD
From time to time, children with delayed neurodevelopmental progress with or without minor dysmorphic features are identified as having a subtle deletion or duplication; and in most, of course, the karyotypic abnormality will have been the cause of the phenotypic defect. Occasionally, parental
268
•
VA R I A N T S
studies will come up with the surprising result that one parent has the same karyotype and a reinterpretation of probable “harmless variant” may be made. The counselor will need to make sure that this reinterpretation is well understood and that the label of “chromosome abnormality” does not attach to the child and stifle other clinical investigation. Equally, the evolving understanding that some “variants” might actually be truly pathogenic, but of low penetrance (with most carriers unaffected), will oblige a nuanced approach. The counselor will need to keep abreast of the literature regarding the significance of these inherited genomic changes. Cancer. Persons who have variant fragile sites (we all have the common fragile sites) may be advised, if the question is raised, that there is no evidence of an increased risk for cancer (Simmers et al., 1987; Sutherland, 1988; Sutherland and Simmers, 1988). There is emerging evidence that some of the common fragile sites have within them tumor suppressor genes that can be disrupted in some cancers (Arlt et al., 2003). But there is no indication of increased risk for an individual in whom a particular common fragile site may be prominent (in terms of the proportions of metaphases in which it is seen).
17 copy number changes benign variants and changes of unclear clinical relevance
THE MICROSCOPE has long been the classical instrument of the cytogeneticist, and large-scale variation in the human genome has been known since the early days of the discipline. As identified by the examination of banded chromosomes, variation can be normal or abnormal. Normal variation includes rearrangements or visible differences that are seen in the general population, and which are without clinical significance. These include the common pericentric inversion of chromosome 9, and the morphological and staining differences between homologs of the acrocentric short arms, and of the pericentromeric heterochromatin of certain chromosomes, and these have been discussed in the preceding chapter. Abnormal variation includes balanced rearrangements such as inversions, reciprocal translocations, and Robertsonian translocations, and unbalanced rearrangements such as derivative chromosomes and supernumerary marker chromosomes. Thus, the traditional study of human chromosomes with banding techniques has identified
large-scale genomic changes, typically referred to as chromosome abnormalities, for over 40 years. The new instrument of the twenty-first century cytogeneticist is the microarray (as described in detail in Chapter 2). This new approach has revealed a new layer of variation: genomic variation ranging from a few to several million base pairs of DNA, in clinically normal individuals. The interpretation of such variation can be that these are normal, abnormal, or of unclear clinical significance. This chapter will explore normal genomic variation and discuss when the lines blur between normal and clinically abnormal, as well as what tools are useful for distinguishing this variation.
MICROARRAY ANALYSIS Chromosome microscopy can identify deletions and rearrangements that involve chromosomal segments greater than 5–10 Mb of DNA and that typically result in an obvious change in banding pattern.
•
269
Alterations smaller than 5 Mb may require molecular methods to visualize. The microscope-based method of fluorescence in situ hybridization (FISH) requires knowledge of where to go “FISHing” to identify a chromosome abnormality. With the advent of microarrays, the entire genome can be scanned for alterations in a single hybridization. Recently, microarray-based chromosome analysis has overtaken traditional chromosome analysis and FISH as the laboratory method of choice for identifying chromosome abnormalities. However, this widespread use of microarray analysis has led to the identification of alterations not previously recognized. It is the job of the cytogeneticist to interpret these alterations and to provide helpful information to the referring physician and genetic counselor. It is then the job of the genetic counselor to convey and interpret the information to the family. Microarray analysis is not a stand-alone test in the diagnostic laboratory, because it cannot elucidate the molecular mechanism that caused the DNA gain or loss, and it cannot reveal the type of rearrangement or the resulting abnormal chromosome structure. The identification of the precise chromosome rearrangement is necessary for proper genetic counseling. For example, the identification of an unbalanced translocation in a child means that one or the other parent could be a carrier of a balanced translocation. As discussed at length in this book, balanced translocation carriers may be at substantial risk for having additional children with the unbalanced form of the rearrangement. Therefore, because the information from a microarray is incomplete in regard to the type of rearrangement, microarray results should be confirmed by FISH to visualize the structure of the abnormal chromosome. For some smaller alterations, FISH analysis may not be possible because the size of the FISH probe is larger than the chromosomal alteration. In these instances, quantitative fluorescent polymerase chain reaction (QF-PCR) or multiplex ligation-dependent probe amplification (MLPA) (Chapter 2) can be used to confirm the gain or loss; although neither of these methods uncovers the nature of the rearranged chromosome. It is true to say that microarray analysis has completely changed the face of cytogenetics and has enabled the detection of chromosome
abnormalities at an unprecedented resolution, and in a considerably greater fraction of patients. DE F I N I T I O N S
Since the identification of the first genomic alterations in normal populations, the literature has been inundated with new terms and attempts at nomenclature, in order to describe the gains and losses observed from microarray analysis. It may be helpful to define the terms as will be used in this chapter. Copy Number Variation. Because cytogeneticists typically refer to “variation” as meaning “normal,” the word variation, or the expression “copy number variation” (CNV), will be used to indicate a benign change in the genome that does not (to the best of present knowledge1) have any phenotypic effect. It is to be seen as a normal observation. (In these early years of the twenty-first century, the broader concept of the CNV is generating a very large literature, well beyond the narrow confines of its relevance to clinical cytogenetics, and we leave this to the interested reader to pursue.) Copy Number Change. On first discovery, the significance of a difference in copy number may not be understood. In general, and if we imagine a “standard genome,” most regions of the genome are represented in two copies, one on each chromosome, albeit that there are some instances of four or more copies of a gene, usually in tandem or near to the “original” copy, with two or more copies on each chromosome, in normal individuals. Thus, copy number change (CNC) refers to a change that is not yet determined to be benign, or clinically relevant with phenotypic consequence. It is simply a gain or loss in which the inheritance may or may not be known. A copy number alteration (CNA) that has clear phenotypic consequence to the individual is not normal: it is a chromosome abnormality and does not have a place in this chapter. Normal Results by Microarray. Such a report indicates that there is no DNA gain or loss of clinical relevance to the phenotype of the individual. The individual may show one or more CNVs, but they are not believed to be relevant to the phenotype. This normal result does not exclude a mutation in a gene that is not detectable by microarray analysis,
1 We can certainly expect advances in knowledge, and surely some CNVs will graduate to the status of pathogenic deletion or duplication. One example from many: a duplication close to the PMP22 gene on chromosome 17p may influence its activity and lead to the syndrome of hereditary pressure-sensitive palsy (p. 323) (Weterman et al., 2010).
270
•
VA R I A N T S
and neither are balanced rearrangements such as reciprocal translocations and inversions excluded, these not being detectable by microarray. Abnormal Results by Microarray. Such a report indicates that there is a DNA gain or loss that is presumed to be causative of the patient’s phenotype. This determination is usually based on the laboratory’s experience with other such patients, with one of the evolving CNV databases, and the available literature on a particular alteration. This determination cannot always be based on size of the alteration or genetic content alone, as some fairly large CNVs (1–2 Mb) are being recognized. Incidental Discovery of Whole Gene Duplication or Deletion. A microarray analysis that interrogates the whole genome may reveal a missing or an extra copy of a gene, quite unrelated to the clinical reason for the test having been ordered. Many genes are not dosage-sensitive in the duplicated state, and for example the incidental discovery of a duplication of the KAL1 gene on the X chromosome would not have any clinical implication (whereas mutation or deletion leads to Kallmann syndrome). However, duplication of PMP22 is certainly pathogenic, and it is the basis of one form of Charcot-Marie-Tooth disease (p. 330); and we refer on p. 16 to the incidental discovery of a duplication that has been associated with an ataxic syndrome. Deletion of a whole gene that is dosage-sensitive or subject to haplo-insufficiency would be pathogenic in the context of dominantly inherited disease; for example, a few cases of the cancer-predisposition syndrome hereditary nonpolyposis colon cancer are due to complete deletions of the MLH1 gene, or of the MSH2 gene. Deletion of one copy of a recessive gene (in the presence of a normal allele on the other homolog) would merely lead to recognition of the carrier state in that individual. We have seen, for example, single-copy deletion of the NPHP1 locus at 2q13 in a number of samples referred from Québec, this gene the basis of, remarkably, both a renal disease (juvenile nephronophthisis) and a cerebellar disorder ( Joubert syndrome). Disease would of course result if both copies were deleted, or if the other chromosome happened to carry a typical mendelian mutation. The ethical issues raised by these discoveries are discussed on p. 15. Results of Unclear Clinical Significance. Rather frequently the microarray analysis reveals a DNA copy number gain or loss, but it is unclear whether this alteration is responsible for the patient’s phenotype.
The laboratory or physician may be unable to assign a clinical relevance to the alteration because it has never been seen before in that laboratory and does not appear in the available literature. Parental testing can be helpful in some cases, as discussed in more detail later. Briefly, if neither parent carries the alteration and paternity is known to be correct, then the gain or loss is de novo and could well be contributing to the phenotype; but equally one must exercise caution, since de novo benign CNVs may also arise (Sharp, 2009). But if one parent carries the alteration and clinical information is not available, or if the parent is clinically abnormal but not necessarily with the same symptoms as the child, the ability to interpret the DNA change becomes more difficult. More specific examples are given later regarding recent evidence that some alterations appear more frequently in the patient populations being studied for a variety of medical issues than in the general, normal population. However, most often in the abnormal patient population, these alterations are inherited from a clinically normal parent. These particular alterations may confer a predisposition to medical problems (a “susceptibility locus”) or represent a modifier to some other, unknown, alteration. It is unclear how many of these types of alterations exist in the population, and this becomes very challenging when encountered in the prenatal setting. As Friedman et al. (2009) rather soberly comment, “it seems likely . . . that no perfect array genomic hybridization platform for detection of pathogenic CNVs may ever exist and that effective clinical interpretation of these studies will continue to require considerable skill and experience.” N OR M A L PO PUL AT I O N VA R I A N TS
CNVs, as defined in this chapter, are gains or losses in the genome that are found among individuals in the general, normal population. The frequencies of each may vary depending on the ethnic background of the individuals tested. The sizes can also vary, from less than 1 kb to a few megabases. The finding of a genomic gain or loss in a child with an abnormal phenotype may require the investigation of parental samples to identify whether the alteration was de novo or inherited. The finding of an inherited alteration lessens the likelihood of a causative relationship between the abnormal phenotype and the genomic change, although causation cannot be completely excluded in all cases because of variability in
Copy Number Changes
•
271
penetrance and expressivity2 of some microdeletions and microduplications (Mencarelli et al., 2008; Buysse et al., 2009). The finding of a de novo change is usually interpreted as causative, but the gene content of this genomic segment should be considered in the context of the patient’s phenotype. In the first studies reported of normal individuals, an average of about 12 CNVs per individual was identified (Iafrate et al., 2004; Sebat et al., 2004). Many studies have evaluated the usefulness of microarrays in the detection of chromosome abnormalities in children with disabilities. CNVs and DNA copy number changes of unclear clinical relevance have often been noted, and Table 17–1 summarizes the earliest of these studies in which 100 or more individuals were tested. The differences in reported frequencies between studies may be due to the kind of array used (BAC, high-density oligonucleotide array, or SNP array) and reporting criteria. Reporting criteria vary widely, with some laboratories relying on internal patient databases, external population databases, and gene content, while other laboratories use an arbitrary cutoff based on size. Because the inclusion criteria of the study subjects and the microarray resolutions differ, it is difficult to combine all studies to find an
overall rate of CNVs. Meta-analysis of reported data showed that more than 20% of the genome was interpreted as a CNV (Cooper et al., 2007; Miller et al., 2010). The array platforms that utilize high-density coverage of the entire genome, irrespective of known CNVs or known clinically relevant regions, have shown up to 31 CNVs per individual studied (Friedman et al., 2006). Every individual showed variation, and most showed many different genomic regions displaying gains and losses. This is in sharp contrast to the targeted microarrays, which show as little as 1%–2% CNVs (Shaffer et al., 2006b, 2007a). The design of the microarray, by avoiding known polymorphic CNVs, can minimize the number of cases requiring parental samples to interpret a child’s result, and it lessens the chance of misinterpreting a benign change as a pathogenic alteration and vice versa. RESULTS O F UN CL EA R CL I N I CA L SI G N I F I CA N CE
There are many circumstances in which findings may be of unclear clinical relevance, with respect to an alteration that has not been reported before.
Table 17–1. Detection Rates of Copy Number Variations (CNVs) in Studies of 100 or More Individuals NO. OF MICROARRAY INDIVIDUALS TYPE, RESOLUTION
INHERITED OR BENIGN CNV RATE
REFERENCE
270
12%
Redon et al., 2006
258/78 normal parentsa 6% 2% 3125/100 childrenb >100% 9% 1% 4% 2% 4%
de Vries et al., 2005 Menten et al., 2006 Shaffer et al., 2006 Friedman et al., 2006 Hoyer et al., 2007 Lu et al., 2007 Shaffer et al., 2007 Baris et al., 2007 Baldwin et al., 2008 Pickering et al., 2008
Aff ymetrix GeneChip 500K and BAC whole genome tiling array BAC, 100 kb BAC, 1 Mb SignatureChip, BAC, targeted Aff ymetrix GeneChip, 100K Aff ymetrix GeneChip, 100K BAC, targeted SignatureChip, BAC, targeted SignatureChip, BAC, targeted Oligonucleotide, 500 kb Spectral Genomics, BAC
100 140 1500 100 104 2444 8789 373 207 1151 a
258 alterations were found in 78 of the tested clinically normal parents of 100 children with idiopathic intellectual disability. 3125 CNVs that “probably represent normal polymorphisms” were identified in 100 children with idiopathic intellectual disability.
b
2 Penetrance refers to the proportion of individuals with an imbalance that shows any trait resulting from that imbalance, whereas expressivity refers to the variability in phenotype of those who carry the imbalanced region.
272
•
VA R I A N T S
If de novo, the alteration more likely contributes to the phenotype. If the alteration is inherited, it may remain unclear because clinical information on the parents is not available, the parent may be clinically abnormal, or the CNC is known to be associated with an abnormal phenotype in some individuals, but not all carriers. Thus, the paradigm for distinguishing pathogenic from benign genomic changes has shifted, and one cannot assume that all DNA changes are fully penetrant. Even de novo changes may not all be pathogenic. Although the mutation rate is unknown, de novo, benign CNV formation cannot be excluded (Sharp, 2009). Some studies have suggested that there is a correlation between the size of the alteration and the probability of its being de novo (de Vries et al., 2005). However, in a review of 432 array comparative genome hybridization (CGH) findings, over 37% of CNVs were inherited from a parent (Menten et al., 2006). We have found that studying the parents for duplications does not provide much helpful information in interpreting the results, as most are inherited. For example, of the unclear significance cases, interstitial duplications of 500 kb–1 Mb in size are de novo only 3% of the time (thus, 97% are inherited). Can it be assumed that the high likelihood of an inherited duplication means that the gain is benign?—perhaps so, but uncertainty may remain. Likewise, small deletions are more likely to be inherited than de novo, although many more of the deletions are de novo as compared to equivalent-sized duplications. Deletions of 500 kb–1 Mb in size were de novo in ~30% of cases, while deletions 250 kb–500 kb and less than 250 kb were de novo 20% and 10% of the time, respectively. Thus, the majority of small deletions are inherited. In another approach, we looked at gene content. The interstitial loss of 6–10 genes was de novo in 27% of cases, 3–5 gene deletions were de novo in 20% of cases, and deletions of 2 or fewer genes were de novo in 14% of cases. Thus, even with small deletions, the chance of finding a carrier parent is high; but the finding of transmission from a reportedly normal parent does not necessarily exclude the possibility that the deletion is pathogenic. In addition, there are now examples of DNA CNCs that are associated with highly variable phenotypes, from abnormal with features in common (suggestive of a syndrome), abnormal with varying features, to a completely normal phenotype in carrier parents. These regions may represent susceptibility loci, but the conditions needed for a penetrant
phenotype are unknown. To illustrate, we use deletion of 1q21.1 and deletion of 16p11.2 as examples. Duplications of these regions exist as well, but the phenotypes are less well characterized than those due to the deletions. Deletions of 1q21.1, of 200 kb extent, are a necessary but not sufficient cause of the thrombocytopenia-absent radius (TAR) syndrome; it is proposed that some other genetic “modifier,” elsewhere in the genome, must coexist (Klopocki et al., 2007). In 75% of cases, the deletions are inherited from a clinically normal parent. In a screen of 5218 individuals with abnormal phenotypes (Mefford et al., 2008), 25 such deletions were identified; the phenotypes were quite variable, including mild-to-moderate mental retardation, microcephaly, cardiac abnormalities, and cataracts. Although not all parents were tested, six deletions were inherited from a clinically normal parent. This deletion was not found in a series of 4373 control individuals. Deletions of 16p11.2 (not to be confused with the cytogenetically visible variant, p. 265), which in some publications have been described also as a CNV, have been associated with autism, as this abnormality appears to be enriched in that population (Weiss et al., 2008; Kumar et al., 2008). However, since this initial observation, several papers have shown that this microdeletion can be found in a wide variety of phenotypes (Bijlsma et al., 2009; Shinawi et al., 2010; Rosenfeld et al., 2010; Bochukova et al., 2010). Phenotypes include speech/language delay, cognitive impairment, motor delay, seizures, poor behavior, and obesity. A clear picture is yet to be drawn (and see also p. 321).
GENETIC COUNSELING The identification of chromosome abnormalities provides family members with an explanation for their child’s developmental disability or birth defect, will allow for better information about recurrence risks, and will permit the anticipation of certain medical problems that may require intervention. The identification of genes causing the clinical features of chromosomal disorders may, in the future, lead to specific treatments. Microarray analysis increases the detection of clinically relevant DNA alterations in patients with developmental disabilities. The improved detection with this new technology needs to be balanced with the detection of results of unknown clinical relevance. Misinterpretation of CNCs as clinically causative, or the inability to Copy Number Changes
•
273
provide explanation to parents of a finding of unclear clinical relevance, is not helpful and may be harmful if the family stops searching for the real answer to their child’s problems. P RE NATAL D IAG N OSI S
Microarray analysis is gaining popularity in prenatal testing, and recent publications have demonstrated the benefits over traditional cytogenetic testing (Sahoo et al., 2006; Shaffer et al., 2008; Tyreman et al., 2009; Van den Veyver et al., 2009; Coppinger et al., 2009a; Faas et al., 2010). The use of microarrays in prenatal testing was recently addressed by the American College of Obstetricians and Gynecologists, which endorsed the use of targeted arrays as an adjunct to conventional cytogenetics in pregnancies with abnormal ultrasound findings (ACOG Opinion, 2009). For this purpose, microarrays should be specifically designed to identify known chromosomal syndromes, which minimizes the detection of CNCs of unclear clinical significance. Prior to prenatal array-CGH, parents should receive comprehensive genetic counseling, including discussion of the inherent limitations of the test. The parents should be told the frequency of unclear results with the particular microarray that will be used, and they should be alerted to the possibility of needing parental samples for FISH or microarray analysis to aid in the interpretation of their prenatal test result. It needs to be understood by the physician, genetic counselor, and parents, that advances in technology may reveal findings about which little is known: their pregnancy might be identified to have a DNA alteration that has not been described before. In that case, little information can be provided about medical prognosis and the clinical spectrum of the disorder. For this reason, the selection of the array content and platform is a most important
274
•
VA R I A N T S
factor in the prenatal setting. In a study by Tyreman et al. (2009), 35 alterations were detected in 106 prenatal specimens; more than 37% of these were classified as CNCs of unclear clinical relevance, using a high-resolution SNP array. In contrast, microarrays designed for the purpose of prenatal testing should have far fewer unclear results (0.5%: Shaffer et al., 2008; Coppinger et al., 2009a). SNP-based arrays can reveal consanguinity or even incest, if unusually high levels of homozygosity are observed, and the family seeking array testing by this method should be aware of this (Schaaf et al., 2011). This is not a concern with array-CGH. After the test results are available, the family will likely need additional genetic counseling depending on the findings. The genetic counselor will expect the laboratory to provide them with information about the alteration. Furthermore, there are public databases that can be of help in deciding the clinical relevance of certain CNCs. Examples are DECIPHER (http://decipher.sanger.ac.uk/application/), the Database of Genomic Variants (http:// projects.tcag.ca/variation/), the database at Children’s Hospital of Philadelphia (Shaikh et al., 2009), and the European Cytogeneticists Association Register of Unbalanced Chromosome Aberrations (Feenstra et al., 2006; ECARUCA, http://agserver01.azn. nl:8080/ecaruca/ecaruca.jsp) and the Signature Genomics Genoglyphix Chromosome Aberration Database (http://www.genoglyphix.com). In the specific case of the 1q21.1 deletion associated with TAR syndrome (see earlier), demonstration of the cytogenetic defect can crucially inform counseling, our incomplete knowledge of the molecular pathogenesis not withstanding, as Uhrig et al. (2007) recount in being able to provide a clear diagnosis in a case of upper limb anomalies detected at fetal ultrasonography.
PART FOUR NORMAL PARENTS WITH A CHROMOSOMALLY ABNORMAL CHILD
This page intentionally left blank
18 down syndrome, other full aneuploidies, and polyploidy
IN THIS CHAPTER we consider the case of parents, themselves karyotypically normal, who have had a child, or a pregnancy that aborted, with a full aneuploidy or a polyploidy. Thus, we include the major trisomies (13, 18, 21) and sex chromosome aneuploidies (XXX, XXY, XYY, and 45,X) as well as less commonly seen autosomal aneuploidies and sex chromosome polysomies. The category of polyploidy is substantially devoted to triploidy. In the great majority, these defects arise from an abnormal event during meiosis or (in some triploidy) at conception. In a few, there is postzygotic generation of aneuploidy. Only in the case of parental gonadal mosaicism, or in the hypothetical setting of an apparent predisposition to meiotic error, will there apply an increased risk of recurrence of aneuploidy, over and above that associated with any parental age effect. Triploidy needs separate consideration.
BIOLOGY Full aneuploidy is presumed in the great majority to be the result of meiotic nondisjunction. A diminished degree of meiotic recombination is typically observed, and this has led Hassold and Sherman (2000) to propose a two-hit sequence, the first hit being a less well-tethered bivalent at meiosis I, and the second hit being a consequential aberrant distribution at meiotic metaphase. Meiotic nondisjunction can happen at any parental age, but it is more frequent in older mothers. Alternatively, an abnormality has arisen in a premeiotic gametocyte, with the parent thus having a “wedge” of gonad that carries the abnormality (gonadal mosaicism). Such a parent would, of course, have an increased risk for only the one karyotypic defect. Finally, a small fraction of apparent full aneuploidy may be due to early
•
277
mitotic nondisjunction in an initially 46,N conceptus with loss, or restriction to extra-embryonic tissue, of the normal cell line. For convenience, we note here also those forms of Down syndrome that are due to translocations (in most of which, however, the genetic imbalance is essentially the same as in the case of standard trisomy).
AUTOSOMAL TRISOMY Trisomy 21 (Down Syndrome) Down syndrome (DS) is the archetypal chromosome disorder. It was the first medical condition shown to result from a chromosome abnormality, in 1959. It has for many years been recognized as the most common single known cause of intellectual disability, and it has the highest incidence at birth of any chromosome abnormality. Every counselor can expect frequently to deal with problems relating to DS and thus should be familiar with its genetics. The Genotype to the Phenotype. The DS phenotype—the characteristic facial appearance, body build, and mental defect—is due, in sum, to a triple amount of chromosome 21. Epstein (2002) provides a broad philosophical and historical as well as scientific review of the central role of the dosage effect in understanding the pathogenesis of the DS phenotype. Particular organ systems are particularly vulnerable, and Torfs and Christianson (1998) have identified characteristic malformations in a population study of nearly 3000 affected infants (Table 18–1). It is, in a sense, a “contiguous gene syndrome,” in which there is an additional dose of an en bloc set of genes. The entire chromosome was sequenced by 2000, and the gene complement turned out to be surprisingly low, only 225 protein-coding loci in all (Hattori et al., 2000). This gene sparseness is plausibly a factor in the survivability of the trisomic state. It was logical that attempts be made to define those regions of the chromosome that might contribute predominantly to the DS phenotype: that is to say, to identify a “DS critical region” (DSCR), which might contain particular “DS genes.” Such an attempt is illustrated in Figure 18–1, with the region 21q22 having the predominant influence. The segment determining the facial features has been narrowed down by study of cases with partial duplications (Kondo et al., 2006), and two rather informative families are those described in Ronan et al. (2007) and in Eggermann et al. (2010b). In the 278
•
Table 18–1. Some Malformations Frequently Observed in Down Syndrome MALFORMATION
RELATIVE RISK
Atrioventricular canal defect* Annular pancreas Duodenal atresia Patent ductus arteriosus* Small intestinal atresia/stenosis Ventricular septal defect* Tricuspid valve defect* Hypoplastic aorta* Tetralogy of Fallot* Atrial septal defect* Ectopic anus Cataract Intestinal malrotation Anal atresia/stenosis Tracheo-esophageal fistula Syndactyly
1009 430 265 152 142 95 84 77 77 71 67 54 45 34 26 26
*Cardiovascular defect. Source: Data from a population study in California 1983–1993, involving 2894 infants with Down syndrome (Torfs and Christianson, 1998).
former, a familial 3 Mb duplication in 21q22.13– q22.2 was associated with a clinically typical DS facies and a mild intellectual deficiency, while in the latter, a child with a 0.46 Mb duplication in a different part of 21q22 had no facial appearance to suggest DS (although he did present a Silver-Russell phenotype). Pritchard and Kola (1999) propose that there are only a few loci whose 150% amount is central in the pathogenesis of DS, and that certain loci may make specific contributions to certain components of the phenotype. One dosage-sensitive gene for which a case has been made is DSCR1, whose very name indicates the presumption of a role. Overexpression of this gene in a mouse reduces the size of the hippocampus, and the number of dendritic spines in certain neurons within this part of the brain; and a smaller hippocampal formation, and fewer dendritic spines, are seen in DS (Keating et al., 2008). And yet the duplication reported in Ronan et al., as mentioned earlier, did not include DSCR1, while that in Eggermann et al. did. DSCR1 is one of a number of “DSCR” loci; each may have their individual contribution. The neural cell adhesion
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
FIGURE 18–1 Phenotypic (trisomic) map of chromosome 21. Thick lines represent regions that must be trisomic to produce the particular trait. Thin-line regions may also contribute to that trait; the contribution of dotted-line regions is less clear. P, profound; M, mild. (From J. R. Korenberg et al., 1994, Down syndrome phenotypes: the consequences of chromosomal imbalance, Proceedings of the National Academy of Sciences of the United States of America 91:4997–5001. Courtesy J. R. Korenberg, and with the permission of the National Academy of Sciences.)
molecule DSCAM, which maps to 21q22.2-q22.3, is strongly expressed (in the mouse) in those equivalent regions of the brain that are compromised in DS, and this is an attractive candidate as a contributor to the DS brain phenotype (Barlow et al., 2001). Another mouse model is pointing to the genes Olig1 and Olig2, mapping to 21q22.11, as candidates; these genes, in the 150% state, may lead to the overproduction of certain brain inhibitory interneurons (Chakrabarti et al., 2010). Duplication of the APP locus at 21q21 is presumed to be the cause of the early-onset dementia that is typically seen in DS, the consequence of a continuing APP overexpression, and hence an overproduction of β-amyloid, which is then deposited in Alzheimer-like plaques in the brain (Bird, 2008). This interpretation is well supported by the observations in the rare form of familial Alzheimer disease due to 21q21 duplication as an isolated genomic rearrangement (the duplications of size range 0.6–6 Mb) (Cabrejo et al., 2006). Other chromosome 21 genes may be of less critical effect when they are overfunctioning, and for some, perhaps many, it may make no difference at all. Shapiro (1997) puts a somewhat different viewpoint, championing the “amplified developmental instability” hypothesis, and comments that “the search for a minimal region on chromosome 21 (the socalled DS critical region) responsible for producing DS has come full circle back to almost the entire chromosome.” In his view, a direct role for one or a few single loci with a one-on-one gene-to-phenotype relationship is simplistic: “traits that characterize DS are complex, and should be viewed and analyzed accordingly.” His general proposition is not unreasonable: that an excess of chromosome 21 encoded gene products perturbs the functioning of the products of many loci, from all chromosomes, in all manner of developmental and physiological pathways. Attempting to draw together the two viewpoints, the gene dosage theory and the amplified developmental instability theory, as do Neri and Opitz1 (2009), we could suppose that the important genetic segments—the “DS loci”—may have their pathogenic role in the modulation, direct or epigenetic (Aït Yahya-Graison et al., 2007), of layer upon layer upon layer of cellular interactions that lead, as
the end result, to a phenotypic range that is clinically recognizable as DS. “Complex” may be too simple a word to describe this. What about the characteristic DS facies? Simply to observe one’s fellows is enough to convince one that development of the human face must be the most subtle and complex and precise process. How could we begin to understand why the DS face is different, and recognizably so? Among others, one contributor may be the failure of the facial musculature to divide into its proper various components in fetal development, and this might have, of itself, a distorting effect upon soft tissue formation of the face (Bersu, 1980). Which gene in triple dose, or which combination of genes, might lead to such a process? Listing the genes that are on chromosome 21, and seeing which ones may or may not be associated with a component of the DS gestalt,2 is merely another step on the way to knowing why this trisomy causes this phenotype. DI FF ER EN T CYTO G EN ET I C F O R MS
The usual basis of DS is standard trisomy 21 (Fig. 18–2). The disorder has a number of other cytogenetic forms, and Figure 18–3 depicts the proportions graphically. Differences in the source and nature of the genetic errors underlying these various forms require each to be considered separately. STA N DA R D T R I S O MY 21 D OW N SYN D R O M E
The great majority (about 95%) of DS is due to simple trisomy of chromosome 21. Around 90% reflects a maternal meiotic error (Yoon et al., 1996). Three-quarters of these maternal errors occur at meiosis I, and one-quarter apparently at meiosis II, albeit that the latter may actually have been set up at meiosis I. Meiotic I errors are associated with reduced or actual absence of recombination between the chromatids of the chromosome 21 tetrad. Particularly an absence of recombination (with no chiasma forming, thus an “achiasmate” tetrad) may lead to each homolog being able to segregate without reference to the other, and thus without the
1 Their review celebrates the fiftieth anniversary of the discovery of the chromosomal cause of DS, and provides a fascinating discussion of nineteenth- and twentieth-century thinking leading up to this event. 2 Gestalt: “a configuration or pattern of elements so unified as a whole that it cannot be described merely as the sum of its parts.”
280
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
1
2
3
6
7
8
13
14
15
19
20
9
4
5
10
11
12
16
17
18
21
22
XX
FIGURE 18–2 Karyotype of a child with standard trisomy 21.
imperative to move symmetrically. It seems probable that the biology of this meiotic susceptibility is universal across the human race (Ghosh et al., 2009). Among the small fraction (about 10%) due to paternal errors, the proportions due to meiotic I and
meiotic II errors are nearly equal. No usefully discernible paternal age effect exists. As in the female, a reduced frequency of recombination observed in the meiotic I cases may underlie the cause of this male nondisjunction (Savage et al., 1998). Two as
4% rob de novo Familial Other Mitotic 1% 25% 75% 3–5% Paternal meiosis 3–5% Meiosis II Meiosis I 75% 25%
St
an
y
Meiosis II 25%
dard trisom
Meiosis I 75%
Maternal meiosis 85–90%
FIGURE 18–3 Origins of trisomy 21 (percentages rounded). Down Syndrome, Other Full Aneuploidies, and Polyploidy •
281
yet unexplained observations concerning trisomy 21 due to paternal meiotic errors are these: this fraction is a little greater among prenatally (11%) than postnatally (7%) diagnosed cases; and there is an excess of males among the DS offspring (Muller et al., 2000). Standard trisomy DS typically occurs as a sporadic event, and recurrences are rare. These categories of cause of recurrence can be listed: gonadal mosaicism, a parental predisposition to nondisjunction, and chance. Recurrence due to Mosaicism. A trisomy 21 cell population in a parent (gonadal, or somaticgonadal mosaicism) is presumed to be an uncommon cause of the production of disomic 21 gametes, although perhaps less rare than originally thought (Bruyère et al., 2000; Mahmood et al., 2000; Kuo, 2002). Pangalos et al. (1992b) studied 22 families in which trisomy 21 had occurred more than once (in siblings, in second- and in third-degree relatives), applying DNA polymorphism analysis. Parental gonadal mosaicism was proposed as the cause of sibling recurrence in 5 of 13 families (about 40%); but other than these, chance alone was enough to explain the recurrences in most, if not all families. James et al. (1998) studied four women, each of whom had had three trisomy 21 conceptions. Two of the mothers were under age 35 at the time of the trisomic conceptions, and they both showed a very low-level mosaicism (0.5% and 4% on blood karyotyping). Neither had a DS phenotype. One of these mosaic mothers was shown herself to have originated as a trisomic conceptus, but an early mitotic loss of a chromosome 21 almost entirely corrected the karyotype. The other two women were older, and in their case it was more likely that the multiple trisomic conceptions occurred independently, as maternal age-related events. In their collaborative series from six Japanese clinics, Uehara et al. (1999c) record the exceptional case of a couple having had five successive pregnancies with trisomy 21 (one DS child, four diagnoses at amniocentesis). Both parents had normal karyotypes on blood and skin analysis. It would seem rather probable that one parent may have had fully trisomic gonadal tissue. Sachs et al. (1990) followed 1211 pregnancies at prenatal diagnosis, subsequent to the occurrence of trisomy 21
in a previous pregnancy, and observed six recurrences (for a rate of 0.5%). In two of these instances, mosaicism was shown. One father karyotyped as 47,+21/46,N on skin analysis; and one mother showed trisomic cells in 3%, 14%, 44%, and 47% on culture of, respectively, blood and skin, and—in a more direct observation—of each ovary. Ovarian biopsy proved the point in a mother of three DS children (and one normal child) who typed 46,XX on peripheral blood, but in whom 8 out of 20 ovarian cells showed trisomy 21 (Tseng et al., 1994). Other similar examples are on record. Nielsen et al. (1988) report a couple having had six documented pregnancies with standard trisomy 21, and five other unkaryotyped pregnancies ending in neonatal death or abortion. The mother typed 46,XX on peripheral blood, and 46,XX/47,XX,+21 in ovarian somatic cells. (Even if the oöcytes were all or nearly all 47,+21, it remains perplexing that no known 46,N conception occurred.) An in vitro fertilization (IVF) setting enabled analysis of the gametes themselves in a woman studied by Cozzi et al. (1999). She had a normal and a DS child at ages 29 and 32, and then had prenatal diagnoses of trisomy 21 at 32 and 36 years. No trisomic mosaicism was detected on peripheral lymphocyte analysis. At IVF, of seven embryos, four were trisomy 21 and one tetrasomy 21, with only two showing normal disomy 21. Four unfertilized oöcytes were analyzed, and three had a supernumerary chromosome 21.3 A rather elegant demonstration of maternal gonadal mosaicism is described in Cupisti et al. (2003), who, in the study of a woman presenting for fertility treatment, identified three oöcyte-polar body pairs having one copy of chromosome 21 in the egg, and two copies in the first polar body. As for the male, Hixon et al. (1998) analyzed sperm samples from 10 men who had fathered a DS child, the additional chromosome 21 having been demonstrated to be of paternal origin. None showed any increase in the fraction of sperm with disomy 21. Recurrence due to Nondisjunctional Tendency. Do some (nonmosaic) individuals, for a certain biological or environmental reason, run an increased risk of producing a trisomic 21 conception? Could a specific sequence within chromosome 21 influence its disjunction (Gair et al., 2005)? Are some people
3 Two of the no. 21 chromosomes had identical haplotypes, indicating that the mother’s mosaicism was due to postzygotic error in an initially normal 46,XX conception (Fig. 3–8a).
282
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
susceptible to a dietary deficiency? Is there a range of “meiotic robustness” in the population? These are perfectly respectable concepts, albeit that they remain quite hypothetical. If so, what possibilities might there be? Several theories for a general predisposition to aneuploidy have been put forward, and some of these are discussed on p. 57. While some of these various possibilities may be more plausible than others, they are all speculative, and we conclude that there is at present no routinely practicable basis enabling the counselor to identify, ahead of time, those parents whose risk is high, and those whose risk is low, to have a second pregnancy with trisomy 21. Recurrence Risk Estimates after One Affected Child or Pregnancy. The earliest estimates of risk are due to Penrose (1956),4 prior to the discovery of the chromosomal basis of DS, and to Stene (1970). Penrose proposed the risk of recurrence to be “doubled, or perhaps nearly trebled” compared to the general population risk, irrespective of maternal age; while Stene derived a figure of 1% for mothers under age 30, with no increase in the age-specific risk for those over 30, at the time of birth of their DS child. More sophisticated analyses were subsequently enabled by the collection of amniocentesis data, and from population studies; and Warburton et al. (2004), Morris et al. (2005b), and De Souza et al. (2009) have determined estimates on the basis of extensive data sets. The advice set out later in the “Genetic Counseling” section is based upon this work. It does remain true that for younger mothers the recurrence risk is, in absolute terms, small. Recurrence Risk Estimates after Two Affected Children/Pregnancies. When a couple have had two (or more) trisomic 21 conceptions, one has to assume an increased risk applies to a subsequent pregnancy, quite possibly a “substantial” risk. The recurrence may well have been due to gonadal mosaicism. Occurrence Risk Estimates with Down Syndrome in a Second- or Third-Degree Relative. More widely in the family, it appears that a history of standard trisomy DS in second- or third-degree relatives does not, in the main, imply an increased risk (Hook, 1992; Pangalos et al., 1992b). Berr et al.
(1990) assessed 188 families in which a DS child had been born, and there were comparable numbers of DS cases among the second- and third-degree relatives, and in the relatives of 185 control families. M OSA I C D OW N SYN D R O M E
47,+21/46,N mosaicism accounts for about 2% of individuals with clinically diagnosed DS. Mosaicism results from a malsegregation of homologs, or an anaphase lag of one homolog, occurring postzygotically. Some individuals with mosaic DS arise from initially trisomic 21 zygotes, losing one of the chromosomes 21 by anaphase lag (Fig. 3–8c in Chapter 3). Others may arise from normal conceptuses, with nondisjunction producing 45,–21/46,N/47,+21 mosaicism, with the 45,–21 line thereafter lost (Fig. 3–8a). Pangalos et al. (1994) studied 17 families in which there was a child with mosaic trisomy 21, and 10 children had three chromosome 21 alleles, indicating their origin from a trisomic conceptus. The chromosome 21, which was subsequently lost to enable formation of the 46,N cell line, showed no predilection for being a maternal or paternal homolog. The remaining seven mosaics had no evidence of a “third allele,” and distinction in these between an initially 46,N or 47,+21 conception was not possible. Whatever the basis, for practical purposes counseling needs to proceed as though the child has standard trisomy 21, recognizing that this will overestimate the risk in some. Genetic counseling for the mosaic individuals themselves is covered on p. 234. I SO CH R O M OS O M E 2 1 D OW N SYN D R O M E
After standard trisomy 21, this is the most common chromosomal category of DS. It has often been called a “21q21q Robertsonian translocation,” but in fact the two 21q components are usually identical, from the same parent, and thus isochromosome is the more accurate term, and the karyotype is more accurately 46,i(21q) (Robinson et al., 1994; RuizCasares et al., 2001). In one series of 112 de novo “rob(21q21q)” probands, none of 130 full sibs and 34 half-sibs had DS (Steinberg et al., 1984). Nevertheless, three of the parents actually showed a low-grade mosaicism, and presumably their having had an affected child reflected that the 21q21q cell
4 His paper was entitled “Some Notes on Heredity Counselling,” and he also referred to “genetical counselling,” one of the first uses of this expression.
Down Syndrome, Other Full Aneuploidies, and Polyploidy •
283
line was included in the gonad. Indeed, a few examples of recurrence in subsequently born siblings are recorded, and parental gonadal mosaicism is the presumed basis of such recurrence (Sachs et al., 1990; Kovaleva and Shaffer, 2003; Chen et al., 2009). This is a point that Mark et al. (1977) directly proved in one case: a woman having sequential pregnancies with the karyotype 46,i(21q) herself typed 46,XX/46,XX,i(21q) on ovarian fibroblast analysis (but 46,XX on blood). Hall (1985) offers the cautionary story of a mother given a low risk of recurrence, who went on to have a second affected child from a second marriage (on resampling of her, a single 46,XX,rob(21q21q) cell was found in 100 cells analyzed). Given this desirability of distinguishing between postzygotic and meiotic (gonadal mosaic) mechanisms, with their differing counseling implications, Kovaleva and Shaffer (2003) advocate study with polymorphic markers in this (and other) homologous Robertsonian translocations. ROBE RTS O N IA N TRA N SLOC AT I ON D OWN SY N D RO ME
Almost all translocation DS concerns a Robertsonian translocation (discussed in detail in Chapter 7). About one-quarter of Robertsonian translocation DS is familial and three-quarters is de novo (1% and 3% of all DS, respectively). De Novo Robertsonian Translocation Down Syndrome. Both parents, by definition, have normal chromosomes. The abnormal chromosome may usually arise as a sporadic event in maternal meiosis I, from a chromatid translocation (Petersen et al., 1991). Such mutational events are rare and, in the great majority of families, recurrences are not seen. But gonadal mosaicism remains a possibility. The so-called rob(21q21q) is, in most cases at least, actually an isochromosome (see earlier). Familial Robertsonian Translocation Down Syndrome. One or the other parent (almost always the mother) is a translocation heterozygote and has transmitted the translocation, in an unbalanced state, to the DS offspring. D OWN SY N D RO ME WI T H RECI PROC A L TRANSLO C ATIO N
The DS phenotype is substantially due, as we noted earlier, to a duplication of the chromosome segment 284
•
21q22.2-q22.3. A reciprocal translocation involving chromosome 21 has the potential to produce, in a gamete from the heterozygote, a duplication of the DS critical region, whether from 2:2 or 3:1 meiotic segregation. The unbalanced adjacent-1 karyotype from the t(18q;21q) illustrated in Figure 5–15 (second row) in Chapter 5 is an example. Or interchange trisomy 21 may result (Fig. 5–13 in Chapter 5). These translocation scenarios are extraordinarily rare, the cause of less than 0.1% of DS. Scott et al. (1995) describe a child with DS from a maternal t(12;21)(p13.1;q22.2), and Nadal et al. (1997) and Lee et al. (2005) describe similar cases from a paternal translocation and insertion, respectively. (It is from studies of cases of partial trisomy 21, comparing those with typical DS and those with different phenotypes, that phenotypic maps, as in Figure 18–1, can be drawn; Kondo et al., 2006.) Interchange trisomy 21 was reviewed by Dominguez et al. (2001), with a total of only 23 published families being accumulated. OT H ER CH R O M OS O M A L F O R MS O F DOW N SYN D R O M E
A number of chromosomally distinct forms of DS result from specific structural changes to chromosome 21. (1) The least rare of these is the terminal rearrangement that produces a mirror-image chromosome around the telomeric region (Pfeiffer and Loidl, 1982). The chromosome has two centromeres, one of which is usually inactive, and satellites on both ends. Such chromosomes are always the result of sporadic mutational events, possibly the result of a translocation between sister chromatids (Pangalos et al., 1992a). (2) DS is seen occasionally in association with other aneuploidies, almost always a sex chromosome aneuploidy, such as 48,XYY,+21 and 46,X,+21; this is known as double aneuploidy. It is usually the result of a double event of nondisjunction resulting in one abnormal gamete. Rather less likely is a scenario of separate events in gametogenesis in both parents. “Interchromosomal effect” has been invoked in standard trisomy DS in the setting of a parental karyotypic abnormality not involving chromosome 21 (e.g., a 13;14 Robertsonian translocation or a reciprocal translocation). In other words, might it be
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
that the rob or the rcp in some way perturbed the distribution of the chromosome 21s? The answer in fact seems to be no, with the case for interchromosomal effect remaining tenuous at best, although a possible exception might be the Robertsonian translocation in the setting of oligospermia (see pp. 148, 153).
Trisomies 13 and 18 (Patau Syndrome and Edwards Syndrome) These syndromes are much less frequent than DS (about 1 in 12,000 and 1 in 6000 live births for trisomies 13 and 18, respectively), and both show a maternal age effect. As with trisomy 21, correlative phenotypic mapping allows certain segments of chromosomes 13 and 18 to be implicated in the genesis of certain phenotypic traits observed in these syndromes (Tharapel et al., 1986; Epstein, 1993; Boghosian-Sell et al., 1994). On molecular studies in trisomy 18, over 90% reflect a maternal meiotic nondisjunction. Uniquely, nondisjunction is considered to happen most frequently at the second meiotic division, this division not taking place until the short period of time surrounding the process of fertilization (Bugge et al., 1998), although there was a contrary view from Verlinsky’s group. From the direct analysis of polar bodies, chromosome 18 meiosis I errors outnumbered those in meiosis II (Verlinsky et al., 2001a). In about 90% of trisomy 13, the additional chromosome is of maternal origin, with meiosis I and II equally susceptible (Bugge et al., 2007); in at least some mosaic cases, the causes may be similar ( Jinawath et al., 2011). Recurrence of trisomy 18 had been recorded in one or two single case reports, and one or two instances of recurrence, or none at all, had been seen in earlier prenatal diagnostic series or retrospective surveys (Pauli et al., 1978; Ferguson-Smith, 1983; Stene et al., 1984; Baty et al., 1994; Uehara et al., 1999c). Baty et al., (1994) noted a 39-year-old mother having had prenatal diagnosis of trisomy 18 at age 39, and a liveborn trisomic 13 infant at age 40 years. No case of trisomy 13 recurrence had been recorded. It had seemed, on balance, that no discernible increased recurrence risk existed, with chance and maternal age the main factors. These may still be the main factors: but in Warburton et al.’s (2004) more recent review, a true increased risk, albeit a very small one, has emerged from the analysis of a very large body of prenatal diagnosis data, and this assessment was confirmed by
De Souza et al. (2009). This is dealt with in more detail in the section on “Genetic Counseling.”
Other Autosomal Trisomy It is extremely rare for any other autosomal trisomy to survive through to (or near to) term. About two dozen examples of each of trisomy 9 and 22 are known, and nonmosaic trisomies 7, 8, 10, and 14 are represented by only one or two reports (Schinzel, 2001; Brizot et al., 2001; Tinkle et al., 2003; Póvoa et al., 2008). In contrast, trisomies are very common in miscarrying pregnancies, a matter dwelt upon in detail in Chapter 23. Robinson et al. (2001) considered that recurrences of trisomy (even of the same trisomy) in spontaneous abortion might represent no more than a common thing happening again, and this may often be the case. Nonetheless, Warburton et al.’s large review (2004) comprised sufficient data to demonstrate that a previously karyotyped trisomic spontaneous abortion indeed does marginally increase the risk for a potentially viable trisomy at a subsequent prenatal diagnosis (see “Genetic Counseling” section), thus indicating, in some instances at least, that an individual predisposition may have been the cause.
Autosomal Monosomy Many autosomal monosomies are presumed to end in arrested growth in the first few mitoses, prior even to the time of implantation, with some possibly proceeding to the stage of “occult abortion” (p. 382), their existence unknown had it not been for the window of observation afforded by preimplantation diagnosis. The single exception may be monosomy 21, albeit that most earlier reports of monosomy 21 have since been reinterpreted as being due, for the most part, to an unbalanced translocation involving chromosome 21 (Cardoso et al., 2008). One presumed case was identified at 17 weeks of pregnancy, going on to fetal death in utero early in the third trimester, although again the cytogenetic diagnosis was not beyond doubt (Chang et al., 2001; Phelan, 2002).
SEX CHROMOSOME ANEUPLOIDY XXY (Klinefelter Syndrome), XXX, XYY These aneuploidies occur at roughly similar frequencies, about 1 per 1000 of the appropriate sex.
Down Syndrome, Other Full Aneuploidies, and Polyploidy •
285
About 75% of XXX and about 40% of XXY Klinefelter syndrome (KS) is due to a maternal meiotic error, and in three-quarters of each of these it is the first meiotic (MI) division that is involved, this MI group showing a maternal age effect. It is noteworthy that almost half of KS results from a paternal MI error (MacDonald et al., 1994). Fathers of paternally originating KS may have marginally elevated levels of disomic XY sperm in comparison with fathers of maternally originating cases, possibly reflecting an inherent tendency among a small minority of these men to produce aneuploid sperm (Eskenazi et al., 2002). In what may have been the only known example of a recurrence, Woods et al. (1997) report two XXY brothers. The karyotype in both reflected a paternal meiosis I error. Manifestly, XYY of meiotic origin must be due to a paternal error, at MII. All three sex chromosomes aneuploidies can have a postzygotic mitotic generation, which may present as mosaicism.
45,X Turner Syndrome In about three-quarters of TS it is the paternal X chromosome that is absent (Hassold et al., 1990a; Uematsu et al., 2002). Mostly, the error is a meiotic one and resides in paternal gametogenesis, possibly reflecting an absence of pairing along most of the X-Y bivalent with a consequential vulnerability in the process of disjunction ( Jacobs et al., 1997). Fathers of nonmosaic 45,Xm Turner girls may have a marginally increased risk to produce sperm nullisomic for a sex chromosome. Martínez-Pasarell et al. (1999) analyzed sperm from four fathers and eight controls, and there was a slight increase in 24,XY sperm (0.22%) and nullisomic sperm (0.48%) in the fathers compared to the fractions in controls (0.11% and 0.32%, respectively). This might suggest that some fathers of nonmosaic 45,Xm Turner girls have a slight proneness to produce sperm nullisomic for a sex chromosome; but if so, the near-absence of recurrences would point to a very minor influence. An alternative explanation is that the loss occurred postzygotically, and the “45,X” child is actually a 45,X/46,XX mosaic, with a very low proportion of XX cells, but this is apparently an uncommon event ( Jacobs et al., 1997). Wiktor and Van Dyke (2005) describe 22 patients with apparently nonmosaic 45,X in whom, upon further study, three
had a minor XX cell line, and 19 were apparently pure 45,X; no XY cell lines were seen. To the contrary, Uematsu et al. (2002) suggest that most TS may actually be due to a structurally abnormal gonosome (X or Y) having been generated in paternal meiosis, with a 46,X,abn(X) conception resulting, and subsequent mitotic loss of the abn(X) leaves a 45,X karyotype. These theories notwithstanding, the observational data point to a very low recurrence risk. In the literature review of Kher et al. (1994), they could find only one instance of 45,X recurrence in sisters. From the Birth Defects Register of Victoria, Australia, over the period 1995–2008, of 245 prenatal diagnoses of 45,X, in none had the indication been of a previous chromosome abnormality ( J. L. Halliday, Murdoch Childrens Research Institute, personal communication, 2010). In the case of a postzygotic origin, if it could be presumed to have been an event that occurred at random in a single mitosis in the early embryo, the risk of recurrence would be regarded not to be raised at all. Kher et al. did however report a unique family with occurrence of 45,X/46,XX in sisters.5 RA R E PO LYS O M I ES
Polysomies such as XXXX, XXYY, XYYY, XXXY, XXXXX, and XXXXY are very rare. Successive nondisjunctions in one parent, the other contributing a single sex chromosome, is the mechanism in most if not all (Hassold et al., 1990; Deng et al., 1991). Apart from the extraordinary circumstance of (hypothetically) a familial tendency to mosaicism, these polysomies arise sporadically (Bergemann, 1962; Kher et al., 1994). Rare reports of coincidence with some other aneuploidy in the family may more likely reflect chance than a causal link (Court Brown et al., 1969).
POLYPLOIDY Triploidy The chromosome count in triploidy is 3n = 69, with a double (2n) chromosomal contribution to the conceptus from one parent (Fig. 18–4). Triploidy can reflect diandry or digyny, with the double contribution coming from the father or mother, respectively
5 A possible role due to the fragile X full mutation in predisposing to X/XX mosaicism is noted on p. 253.
286
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
1
2
3
6
7
8
13
14
15
19
20
9
4
5
10
11
12
16
17
18
21
22
XXY
FIGURE 18–4 Karyotype of a 69,XXY triploid fetus (see also Fig. 23–4 in Chapter 23).
(also referred to as Types I and II triploidy) (Fig. 18–5). The great majority of triploid conceptions abort during the first or early second trimester. Of those aborting at the embryonic stage, most are digynic, while in contrast, most fetal losses reflect a diandric state (McFadden and Robinson, 2006). The appearances on morphological examination at the stage of the embryo do not differ according to a
digynic versus diandric origin, whereas the clinical presentations are readily distinguishable by the fetal stage of development, the latter distinction possibly reflecting a placental-mediated influence. The Two Distinct Forms of Triploidy Diandry is usually, almost always, the consequence of dispermy; that is, two sperm simultaneously fertilizing
FIGURE 18–5 The three major routes whereby triploidy may arise. A complete failure of a meiotic division produces a diploid egg (left) or sperm (middle). Simultaneous fertilization by two sperm is dispermy (right). Down Syndrome, Other Full Aneuploidies, and Polyploidy •
287
the ovum (Zaragoza et al., 2000; McFadden et al., 2002).6 The fundamental problem in this instance may lie in the “zona reaction,” which is the response of the investing shell of the ovum, the zona pellucida, to prevent further sperm entering after the first has penetrated. A minority are due to fertilization with a diploid sperm, this diploid state having arisen from a complete nondisjunction in spermatogenesis. Digyny is most commonly due to a diploid egg, which may be the result of nondisjunction of the entire chromosome set at either the first or the second meiotic division in oögenesis, meiosis II being the more vulnerable, or, of the fertilization of a primary oöcyte7 (Zaragoza et al., 2000; McFadden and Robinson, 2006). A rare cause may be the fusion of two eggs (whimsically called “dieggy”). Individual susceptibility may exist, as discussed later. Diploidy can be presumed to exist in the “giant binucleate oöcyte,” and these visibly abnormal gametes have actually been shown at IVF to lead to a triploid embryo (Balakier et al., 2002; Rosenbusch et al., 2002). Natural History. Triploidy is not uncommon in early pregnancy (1%–3% of recognized conceptions), but about 99.99% are lost as first-trimester miscarriage or second-trimester fetal death in utero. From Hawaiian data, of 38 recognized triploid pregnancies over the period 1986–1999, around 40% were XXX, 60% XXY, and a single case of XYY. Most (80%) aborted early, a few (10%) presented as fetal deaths in utero, and 10% were electively terminated (Forrester and Merz, 2003a). Of all 16-week pregnancies, only 1 in 30,000 are estimated to be triploid, and at 20 weeks, only 1 in 250,000 (Snijders et al., 1995). Diandric triploids mostly abort in the 10- to 20-week period, the mean at 12 weeks. The very few diandric triploid pregnancies that survive to the second trimester typically show partial hydatidiform mole; growth retardation is usual but not invariable (Daniel et al., 2001). Dygynic triploids mostly abort
early (mean 10 weeks), although those exceptional few that remain are able to continue through to the third trimester, when they come to outnumber diandric cases. These surviving digynic triploids develop as a severely growth retarded fetus with marked head-body disproportion, the head being relatively large, and with an abnormally small and nonmolar placenta (McFadden and Langlois, 2000; Daniel et al., 2001). In one case of a digynic 69,XXX triploid coexisting with a normal 46,XY twin, survival to 20 weeks (when selective feticide was done) may have been supported by the normal fetus (Gassner et al., 2003). Intrauterine survival may also be promoted if there is fetal-placental karyotypic discordance, with the placenta being diploid (Kennerknecht et al., 1993a). Survival to the third trimester is associated almost invariably with perinatal death. Of those liveborn, hardly any digynic triploids survive for more than a month; there is one extraordinary instance of death not until 312 days (Sherard et al., 1986; Hasegawa et al., 1999). RECUR R EN CE
While most triploidy occurs sporadically, a genetic predisposition does exist, and recurrences are well described. In one case, the cause was laid at recurring maternal meiosis II errors: a woman coming to IVF, having had two previous triploid pregnancies, had triploidy identified by preimplantation genetic diagnosis (PGD) in 2 out of 13 conceptions (Pergament et al., 2000). In this example, a recurrent maternal meiosis II error8 could be implicated, as might also be so in a similar case in Check et al. (2009): since fertilization had been achieved with a single sperm at intracytoplasmic sperm injection (ICSI), diandry was clearly excluded. More difficult to explain is the occurrence of triploidy of both digynic and diandric bases to the one couple, and the occurrence of both partial hydatidiform mole (due to diandric triploidy) and complete mole (the rare type associated with biparental disomy) to the same
6 Dispermy could be deduced simply from the cytogenetic analysis in the case reported in Lim et al. (2003), the man carrying a translocation 46,XY,t(2;6)(p12; q24). The 69,XXY mole had both the balanced translocation and one unbalanced form, reflecting fertilization with one sperm from alternate segregation and the other from adjacent-1. 7 One extraordinary case was due to the fertilization, by a normal haploid sperm, of a first polar body, which carried a diploid chromosomal set (Bieber et al., 1981). Although acardiac, in utero survival of the triploid fetus was enabled by its normal co-twin, who had arisen from a normal fertilization of the egg. The egg and its first polar body (PB1) had respectively haploid and diploid constitutions as depicted in Figure 3–1 (in Chapter 3), lower right. 8 At the same PGD procedure, 3 of the 13 embryos were trisomic 18, and as noted earlier, this trisomy is mostly due to a meiosis II error.
288
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
couple (Kircheisen et al., 1991; Deveault et al., 2009). A role for the NLRP7 gene is emerging. These aspects are discussed in more detail in Chapter 23. Diploid/Triploid Mosaicism. Van de Laar et al. (2002) accumulated 25 cases from the literature and reported three of their own. These three came from a population catchment of 15 million over a 20-year period, attesting to the rarity of the condition. The triploid line typically reflects digyny, and the basic mechanism may be inclusion of the second polar body at a very early stage after conception of a diploid zygote. Similarly in diandric cases, the mechanism may be dispermy, but with one sperm pronucleus sequestered in the cytoplasm for a few divisions before being incorporated into the nucleus (Daniel et al., 2003b; Wegner et al., 2009). Daniel et al. refer to “delayed digyny” and “delayed dispermy,” respectively, as the course of events whereby the extra pronucleus sits to one side, so to speak, while the diploid lineage is in the process of being established, and the pronucleus then being taken up into the nucleus of one blastomere to give rise to the triploid cell line. Survival of the affected fetus in utero is presumably promoted by the diploid cell line. In most cases the triploid line is not seen on a blood analysis, and fibroblast culture is necessary (Boonen et al., 2011). A single instance of a false-negative amniocentesis is to be noted (Flori et al., 2003). Rare Complexities. “Hypotriploidy” describes the circumstance of a 68-chromosome constitution. The usual mode of formation may be fertilization of a diploid egg with a 22,–X sperm, leading to a 68,XX karyotype; the phenotype resembles that of digynic triploidy (Pasquini et al., 2010). 45,X/69,XXY mosaicism is recorded in a single case, an infant presenting with genital ambiguity, and who displayed complete soft tissue syndactyly of the index and middle fingers of one hand (this being a feature of triploidy) (Quigley et al., 2005). On blood, the karyotype was nonmosaic 45,X, and on skin fibroblast culture, 45,X[3]/69,XXY[77]. The authors propose an initial 46,XY zygote, which lost an X in one cell at possibly the first cell division, giving rise to the 45,X lineage, followed by delayed dispermy of a (or the) 46,XY cell, to give the 69,XXY cell line.
The simplest, and most usual, may be reduplication of the diploid set in the zygote: at the first mitosis, the chromosomes replicate, but the cell fails to divide. These would karyotype as either 92,XXXX or 92,XXYY. Such a mechanism could not explain the very rare case of 92,XXXY, and here it is necessary to invoke such scenarios as retention of a polar body with concomitant dispermy, trispermy, or dispermy with a haploid and a diploid sperm (Baumer et al., 2003; Surti et al., 1986). In one instance, a woman having PGD for recurrence of digynic triploidy had one 92,XXXX embryo, possibly suggesting a meiotic susceptibility in her case (Check et al., 2009). The typical phenotype is that of miscarriage with complete hydatidiform mole, or “hydropic abortion” (Fukunaga, 2004) Tetraploidy in a term pregnancy is exceedingly rare, and survival in one apparently nonmosaic case to 26 months unprecedented (Teyssier et al., 1997; Guc-Scekic et al., 2002). Mosaic diploidy/tetraploidy in a person has been described in association with severe mental defect, and it may only be detectable on skin fibroblast study (Edwards et al., 1994). A complex case is that reported in Leonard and Tomkins (2002) of a retarded woman with body asymmetry and hypomelanosis of Ito, in whom some fibroblasts cultured from hypopigmented skin showed 92,XXXX, others being 46,XX and 46,XX, t(1;6)(p32; q13), and 46,XX on blood. True diploid/tetraploid mosaicism may be quite frequent at the blastocyst stage of development, but either the abnormal embryo is cast off shortly thereafter or, especially if the proportion of tetraploid cells is small and the blastocyst is otherwise of good quality, the polyploid component may be confined to the trophoblast and in due course come to comprise a minor fraction of placenta (Clouston et al., 2002; Bielanska et al., 2002b). Possibly for this reason, tetraploidy can occasionally be seen at chorionic villus sampling (CVS) and at amniocentesis, reflecting a “normal” tetraploidy of part of the placenta, with the remaining extrafetal and fetal tissues being karyotypically normal (Benkhalifa et al., 1993). Alternatively, tetraploidy at prenatal diagnosis may be artifactual.
GENETIC COUNSELING
TE T RAP LO IDY
Down Syndrome
A number of mechanisms may lead to a conceptus with 92 chromosomes, four of each homolog (4n = 92).
The central requirement for accurate genetic advice in DS is knowledge of the chromosomal form in the
Down Syndrome, Other Full Aneuploidies, and Polyploidy •
289
affected family member. If a child diagnosed as having DS has died and no chromosome studies were performed, it may be reasonable to check for the possibility of a familial translocation in the consultand(s). P RE VIO US C H ILD WI T H STA NDA RD TRI SO MY 2 1 (IN C LU DI N G M OSA I C I SM )
If the child has standard trisomy 21, or is a 47,+21/46 mosaic, it is unnecessary routinely to study the parents’ chromosomes. One can assume, with considerable confidence, that they will type as 46,XX and 46,XY. The risks for recurrence of trisomy 21 in a subsequent amniocentesis, or occurrence of a different aneuploidy, are summarized as follows, and as set out in detail in Tables 18–2 and 18–3, and with reference to the studies of Warburton et al. (2004), Morris et al. (2005), and De Souza et al. (2009). • For a mother under 30 years old at the birth of the DS child, and who is still under age 30, the risk
for recurrence of trisomy 21 at amniocentesis is about eight-fold (8.2×) her age-related risk. • If a mother who was under 30 at the birth of the DS child is now over age 30, her current age is the predominant risk factor, and the additional risk due to the previous affected pregnancy is about two-fold (2.2×) her age-related risk. • If a mother was over 30 at the birth of her DS child, the current age-related figure becomes the more important factor, especially as she gets into her later thirties and forties, and the extra risk due to the previous DS child increases her age-related risk only by about one and a half-fold (1.6×) (and see Table 18–3, column B.). • The risk of recurrence at birth will be a little less than at prenatal diagnosis, reflecting a greater likelihood for natural loss of a trisomic pregnancy in the period following the time of amniocentesis. • The risk for a different viable trisomy (amniocentesis diagnosis) is a little above double the age-related risk (2.4×) for younger women under 30 at the previous DS pregnancy, and a
Table 18–2. Increases in Recurrence Risk, Given as Multiples Compared with the Maternal Age-Related Baseline, for Women Who Have Had a Previous Trisomic Pregnancy FOLD INCREASED RISK OF RECURRENCE FOR: PREVIOUS ABNORMAL PREGNANCY
SAME TRISOMY
OTHER VIABLE TRISOMY
Trisomy 21 at maternal age <30 current maternal age <30 Trisomy 21 at maternal age <30, current maternal age ≥30 Trisomy 21 at maternal age <35 Trisomy 21 at maternal age ≥30 Trisomy 21 at maternal age ≥35 Trisomy 13 overall Trisomy 18 overall Trisomy 13 or 18 at maternal age <35 Trisomy 13 or 18 at maternal age ≥35 Trisomies 13, 18, XXX and XXY Nonviable trisomy in spontaneous abortion*
8.2×
2.4×
2.2×
2.4×
3.5× 1.6× 1.7× 8.6–9.5× 1.7–3.1× 7.8× 2.2× 2.3×
1.3× 1.7× 1.5× 1.5× 1× 1.6× 1× 1.6× 1.8×
Notes: Separate figures are given for the risk of recurrence of the same trisomy (“homotrisomy”) or of a different trisomy (“heterotrisomy”). If wished, the appropriate multiple for a particular case can be applied to the woman’s current age-related risk, as listed in Tables 24–2 to 24–5 in Chapter 24, in order to generate an adjusted recurrence risk figure. Figures are from the prenatal data of Warburton et al. (2004) and pre- and postnatal data of De Souza et al. (2009), and they are grouped in various ways, according to the formats of these papers. Specific age-related figures for previous trisomy 21 are also given in Table 18–3, column B. *But cf. Robinson et al. (2001), who discerned no increased risk following an aneuploid miscarriage.
290
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
Table 18–3. Estimates of Recurrence Risk for Trisomy 21, According to the Mother’s Current Age (Column A), and According to Her Age at the Birth of the Affected Child (Column B) A. BASIC AGE-SPECIFIC RISK MATERNAL AGE AT THIS CURRENT PREGNANCY
20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50
RISK (‰)
0.9 0.9 0.9 0.9 0.9 1.0 1.0 1.1 1.1 1.2 1.4 1.6 1.9 2.3 2.9 3.7 4.9 6.6 8.8 11.7 15.2 19.2 23.5 27.8 32.0 35.8 39.2 42.1 44.5 46.4 48.0
B. ADDITIONAL RISK DUE TO PREVIOUS DS AGE AT THE EARLIER DS PREGNANCY
20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50
RISK (‰)
6.2 6.2 6.1 6.0 5.8 5.7 5.4 5.2 4.8 4.4 4.0 3.5 2.9 2.4 1.9 1.5 1.1 0.8 0.6 0.5 0.4 0.3 0.2 0.2 0.2 0.2 0.1 0.1 0.1 0.1 0.1
Notes: Risks A and B are then to be summed. This combined risk figure relates to the probability of detection of trisomy 21 at early second-trimester amniocentesis. For example, a woman who is now pregnant, and due to deliver at age 30 (risk = 1.4‰ from column A), and who had had a DS pregnancy when she was 25 (additional risk = 5.7‰ from column B), has an overall risk for trisomy 21 in the current pregnancy of 1.4 + 5.7 = 7.1‰, or 1 in 141. Note how, with advancing maternal age at the current pregnancy (A), the additional risk component due to having had a previously affected child (B) progressively diminishes; in other words, at these older ages, the maternal-age factor becomes the overwhelming contributor to the risk. DS, Down syndrome; ‰, per 1000 Source: From Morris et al. (2005b).
little below double the age-related risk (1.7×) for those 30 or over at the DS pregnancy. Since the baseline figures are very low (see Table 24–4 in Chapter 24, p. 408), the absolute risk figure is still a low one. • If 35 years, rather than 30, is used as the cutoff age for younger/older mothers, a slightly different perspective is given (Table 18–2). In any event, regardless of the exact figure, the practical point is that the risk for a recurrence of DS is comfortingly low, only approaching the 1% mark by the mid-thirties. Nevertheless, most couples seek the reassurance of prenatal diagnosis in pregnancies after having had a child with DS. Elkins et al. (1986b) observe that some of these parents declare they would not abort a trisomy 21 fetus, and the counselor needs to be sensitive to possible ambivalent feelings of the parents in this setting. T WO PREVIO US TR ISOM I C 21 CO N CEPTIO N S
One can only offer an educated guess that the risk for a third trisomic conception will be “substantial.” A skin biopsy study would be largely academic. If gonadal mosaicism (rather than de novo recurrence) is the cause, a considerable fraction of whichever gonad it is must be involved, since two separate samplings have already come from this fraction. A risk in the range of 10%–20% may be a fair figure to offer. Preimplantation genetic diagnosis would have an obvious attraction. I SOCH RO MOS O ME 21 D OW N SY N D RO ME
From the 0/164 fraction among siblings of de novo isochromosome 21q DS in Steinberg et al.’s series (1984), the risk for recurrence is presumed to be small. Nevertheless, three parents (3%) in this series were demonstrably mosaic, and there are a handful of recurrences otherwise on record; a cautious stance is thus prudent. A risk figure in the region of 1%–2% may be a reasonable one to offer. P RE VIO US C H ILD WI T H ROBE RTSON I A N TRANSLO C ATIO N D OW N SY NDROM E
Obviously, distinction between de novo and familial forms of translocation DS is crucial; this distinction is made by chromosomal studies of the parents. 292
•
For the de novo translocation, a recurrence risk figure of <1% is applicable (Gardner and Veale, 1974). In the case of familial Robertsonian translocation DS, the genetic risk for the female carrier is substantial. The risk to have a liveborn child with translocation DS is about 10%, while the likelihood to detect translocation trisomy 21 at amniocentesis is about 15%. For the male carrier, the risk to have a child with translocation DS is small, about 1% (and see Chapter 7). PREV I O US CH I L D W I T H NONR O B ERTS O N I A N T R A N S LO CAT I O N DOW N SYN D R O M E
In the rare instance that translocation DS is associated with a familial reciprocal translocation, the principles presented in Chapter 5 are to be followed. PREV I O US CH I L D W I T H OT H ER C HR O M OS O M A L CAT EG O R Y O F D OW N SY ND R O M E
For sporadic structural changes such as the terminal rearrangements, the risks are presumed to be very low (less than 0.5%). For the double aneuploidies, there is no evidence to suggest the risks are any different from the recurrence risks for standard trisomic DS. WI D ER FA M I LY H I STO R Y OF D OW N SYN D R O M E
There is no conclusive evidence of an increased risk for second- and third-degree relatives of individuals with standard trisomic DS themselves to have offspring with the condition. The appropriate action in the setting of “a family history of DS” is to determine whether the affected member has standard trisomy 21. If this is so, the family may be reassured that there is no discernibly increased risk, which advice could also reasonably be offered if a single case was associated with older maternal age. If the karyotype of the index case is unknown, and the mother had been younger, the small possibility of a familial translocation may be checked by chromosome study of the counselee. T RI S O MY 2 1 I N PR O D UCTS OF CO N CEPT I O N
The finding of trisomy 21 in products of conception after spontaneous abortion (in those centers where
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
this testing may be done) presents a problem. Should this, for genetic counseling risk assessment, be regarded as equivalent to having had a child with DS? From about 10 weeks gestation through to term, about a third of trisomic 21 conceptions are lost (p. 405), and it may be stochastic events in utero rather than intrinsic genetic differences that distinguish those that abort and those that survive. It may be prudent to err on the side of caution and provide a risk figure as though the abortion had been a liveborn child.
Trisomy 13 and Trisomy 18, and Other Autosomal Trisomy Recurrence of trisomy 13 or 18 is a very rare observation. Nevertheless, in Warburton et al.’s (2004) data, a true increase in risk did emerge, albeit that the absolute figure will still be very small, given the very small baseline age-related figures. In the study of De Souza et al. (2009), based upon 748 women who had had a previous trisomic 13 or 18 pregnancy, a similar increase in risk was observed. The numbers were small (albeit the largest such study ever undertaken), and of 1057 subsequent pregnancies, recurrent trisomy 13 or 18 was diagnosed in 9. Relative risk estimates from these two studies are displayed in Table 18–2. If there is an increased risk for a different potentially viable trisomy (“heterotrisomy”), it must be very small, and Warburton et al. (2004) derive a 1.6fold factor. In De Souza et al. of the 1057 pregnancies just noted, 8 had trisomy 21, but this number was scarcely different from expectation based upon age. In the case of a previous pregnancy with some other type of autosomal trisomy (typically identified in products of conception following spontaneous abortion), from Warburton there may be an increased risk (1.8-fold), but very small in absolute terms, for a potentially viable trisomy in a subsequent pregnancy, although from Robinson et al. (2001), no such risk was discerned. It seems likely that many, if not most cases of recurrent trisomy in older women, whether in spontaneous abortions or in live births, represent the increased risk of a trisomic conception related to maternal age as the key predisposing factor.
XXX, XXY, XYY, 45,X, Other Sex Chromosome Aneuploidy There is no firm evidence that a recurrence risk above the age-specific figure exists, and indeed in
respect of XXX and XXY no recurrences, of either homotrisomy or heterotrisomy, were observed in the study of Warburton et al. (2004). Prenatal diagnosis is discretionary.
Triploidy Diandric triploidy associated with partial hydatidiform mole has a ~1% risk of recurrence; we discuss this in more detail on p. 400. As noted in the “Biology” section, some women may have a predisposition for digynic triploidy. However, the level of risk for recurrence of triploidy, or occurrence of an aneuploidy, must usually be small, since in the series of Robinson et al. (2001) no increased risk was discernible, for women having had more than one previous spontaneous abortion due to triploidy (or aneuploidy), to have yet another chromosomally abnormal pregnancy. Prenatal karyotyping and/or early pregnancy ultrasonography may reasonably be offered. The prevention of chromosomal pathology, as a direct exercise, largely involves secondary prevention: in essence, the selective termination of pregnancies in which a chromosomal abnormality has been identified, or the discarding of abnormal embryos following PGD. Primary prevention is indirect, and encouraging a younger maternal age may be the only feasible approach, absent any clear understanding of environmental factors that might compromise the chromosomal integrity of gamete or zygote. But one remarkable exception to this state of affairs concerns the actual correction of a chromosomally abnormal zygote: and this involves the diandric triploid zygote, otherwise destined to undergo implantation failure or, in the minority that actually implant, to proceed to a severe fetal defect. A triploid zygote due to dispermy will possess three pronuclei. In vitro removal of one pronucleus, at IVF, would restore normality. This would have to be, in the case of dispermy, one of the paternal pronuclei, thus leaving one maternal and one paternal pronucleus. Escribá et al. (2006) applied this approach to tripronuclear embryos in the research laboratory, removing the pronucleus farthest from the second polar body (the one closest to the polar body being very likely maternal), and followed the embryo through to the blastocyst stage. They were able to confirm restoration of diploidy, and could also observe that these corrected embryos showed normal development at day 5, unlike the uncorrected embryos, in which no inner cell mass was seen to
Down Syndrome, Other Full Aneuploidies, and Polyploidy •
293
form. And in the first ever example of “chromosomal cure” of a child-to-be, Kattera and Chen (2003) corrected a tripronuclear zygote, implanted the embryo, and a normal 46,XY boy was subsequently born. These authors comment, cautiously, that this approach should be used “only as a last resort.” In contrast, Pergament (2010) boldly predicts that, by 2020, we will fully understand the mechanisms of meiosis, and we will be able to “treat oöcytes, sperms and preimplantation embryos to ensure that the
294
•
euploid state will be obtained at conception and then maintained during early embryonic development,” initially doing this in vitro, but eventually in vivo. We shall see. T E T R A PLO I DY
True (that is, not artifactual) tetraploidy is too rare for a clear picture to have emerged. Sporadic occurrence would seem very probable.
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
19 structural rearrangements
IN THIS CHAPTER we consider the circumstance of parents, themselves typically karyotypically normal, who have had a child in whom a structural chromosome rearrangement has been identified. Under this heading, we distinguish in particular deletions (partial monosomy) and duplications (partial trisomy). If the rearrangement occurs during meiosis, or at a postzygotic mitosis, we generally assume a recurrence risk no different from the general population. These cases arise anew—de novo—with the affected child. If, however, the rearrangement arises at a premeiotic mitosis, the parent would be a gonadal mosaic, and an increased risk for recurrence, for a second child with the same abnormality, could in theory apply. Usually, no prior distinction between these two possibilities can be made, although how often a rearrangement is observed (unique/nonrecurrent, or commonly seen/recurrent) may suggest the site of generation (see later). Here we consider those deletions and duplications in which cytogenetic or molecular cytogenetic techniques are important in demonstrating the defect, and which
are generally thought of as being chromosomal conditions. In some, and more particularly those now coming to light through the microarray, there is a possibility that a parent might be a carrier, and so our focus is not exclusively on de novo defects. Some of these deletions and duplications occur sufficiently frequently, and/or present a sufficiently distinctive phenotype, that they have acquired syndrome status. The classical route whereby a chromosomal syndrome came to be established followed the recognition of a group of patients with a very similar clinical picture, often with a characteristic dysmorphology: “phenotype-first.” Subsequent cytogenetic studies revealed the underlying chromosomal basis in common (in the case of Down syndrome, this took nearly a century). Nowadays, the typical approach is “genotype-first,” or “reverse dysmorphology” (Shaffer et al., 2007; van Ravenswaaij-Arts and Kleefstra, 2009). Subtle deletions and duplications may not present a distinctive enough phenotype that would allow the clinician to “call” a syndrome. But in the laboratory, recurrent rearrangements, whether •
295
seen in house, or in collaboration with other cytogenetic services, nationally or internationally, can be collected. It is then up to the clinicians to draw together the observations from the patients thus identified and to construct the core features of the new syndrome. This new approach of identifying the chromosomal abnormality first can reveal the natural clinical variation of the genomic rearrangements, which might never have been possible with the traditional phenotype-first approach.
BIOLOGY Mechanisms of Formation of Structural Rearrangement Human chromosomes are disconcertingly dynamic structures. Genomic DNA can be deleted, duplicated, and moved around, with untoward results. Conditions that are the consequence of these rearrangements in underlying genomic structure or architecture may be referred to as “genomic disorders” (Lupski, 2009). Older expressions, not to be discarded, include “partial aneuploidies,” “segmental aneusomies,” and “contiguous gene disorders.” This is in clear contrast to the classic “pure” whole chromosome aneuploidies, in which no rearrangement has taken place. These genomic rearrangements are typically generated from the operation of a chromosomal repair process. Different categories of rearrangement can be considered, according to whether the repair process is homology-dependent or homology-independent; whether the event takes place during meiosis or in a mitotic cell; and whether the recombination is within a chromosome (intrachromosomal) or between chromosomes (interchromosomal). NONALLELIC H O MOLOGOU S RECOMB IN ATIO N
The common basis for many of these rearrangements, and more so the recurring ones, lies in the existence of multiple DNA sequences throughout the genome, generally of some thousands of base pairs, which are sufficiently similar (“paralogous,” rather than exactly homologous) that they enable the erroneous coming-together of different chromosome regions. Within the two sequences (inter- or intrachromosomal) involved in a particular exchange, there is a length of perfect or near-perfect homology, and this is the site of the actual strand exchange (nonallelic homologous recombination [NAHR]). 296
•
Studies on the Charcot-Marie-Tooth region of 17p12, and the Smith-Magenis syndrome region in 17p11.2 (Fig. 19–1), in particular, have informed this work (Lupski, 2009). These sequences, whose misalignment during meiosis sets the stage for the recombination between the two regions, have been dubbed “duplicons,” segmental duplications, or low copy repeats (Gu et al., 2008). Some paralogous sequences run in opposite directions; and in that case, paracentric microinversions, existing as normal polymorphisms, make it possible for this recombination event to take place. The syndromes of duplication or deletion of 15q13.3 (see later) in particular have been well studied in this respect (Sharp et al., 2008). NONH O M O LO G O US EN D JO I N I N G
During mitotic cell division (and including in the premeiotic gametocyte), aberrant DNA replication and repair may lead to the generation of a rearrangement. Different chromosomal segments may happen to be in close proximity, due to their “geographical space” within the nucleus; or upstream or downstream sequences with similarity (such as Alu repeats) may have predisposed to their coming together (this more so in the case of deletions). Then, if breaks occur during replication, instead of the correct ends being brought back together, the broken ends of different segments may inappropriately be ligated. This is nonhomologous end joining (NHEJ). If two segments share a few nucleotide sequences in common (“microhomologous patches”), this may of itself predispose to apposition and recombination: “microhomology-mediated end joining.” Or the active replication fork may “stall” and switch to a template of similar microhomology elsewhere on the same or another chromosome, which happens to be in the same “space” within the nucleus. This is “fork-stalling and template-switching”; and as well as simple deletions and duplications, this may be the basis of some more complex rearrangements, such as triplications, and deletion/duplication combinations (Zhang et al., 2009a). In contrast to NAHR, these mechanisms apply particularly to deletions and duplications seen only in nonrecurring cases (Vissers et al., 2009). ORIG I N PR E- , I N T R A - , O R POST M EI OS I S
As just noted, the recurring de novo deletion or duplication is considered typically to originate at
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
FIGURE 19–1 One mechanism to produce a duplication and a deletion (and see text) is based upon the promiscuous activity of similar DNA sequences, “duplicons,” that exist at numerous places along the chromosome (shown as crosshatched segments). Consider a segment between two such duplicons, indicated here by the black and white spots (imagine these to be two fluorescence in situ hybridization [FISH] probes). Misalignment of the two duplicons, followed by nonallelic homologous recombination within them (×), produces recombinant products that are reciprocally imbalanced: one with a deficiency of the chromatin between the two duplicons, and the other with a duplication. The deletion chromosome is shown with no black or white spot, while the duplication chromosome has a double set of black and white spots in tandem. The general case is drawn after Chandley (1989). The genotype in the child (normal, deletion, duplication) resulting from such meiotic recombination will depend upon which of the chromatids ends up as the homolog in the gamete. The two classic examples both reside in chromosome 17. Smith-Magenis syndrome and Potocki-Lupski syndrome are due to deletion and duplication, respectively, for the segment 17p11.2p11.2. On a smaller scale, and just a little further up 17 short arm, a 1.7 Mb segment within 17p12 including the PMP22 gene is deleted in hereditary pressure-sensitive neuropathy and duplicated in Charcot-Marie-Tooth neuropathy.
meiosis, and the child is nonmosaic. Nonrecurring abnormalities, in contrast, may have arisen at a premeiotic mitosis, such that the parent is a gonadal mosaic, with a “wedge” of the gonad containing the abnormal chromosome constitution (note that the child would be nonmosaic). In fact, a second affected child of such a parent is scarcely ever seen, and one might conclude that any such wedge would comprise only a very small fraction of the gonad. Nevertheless, it is usually appropriate to check the parental karyotypes (as well as for the reason of reassurance), in order to test the possibilities that one parent may be either a carrier of a balanced rearrangement, or a low-grade mosaic for the abnormal
chromosome (the normal cell line in a phenotypically normal parent being, presumably, predominant in the soma). An example is illustrated in Figure 3–12 (in Chapter 3) of an interstitial deletion del(1) (q25q31.2) which was identified at amniocentesis, and which led to the discovery of 46,XY[20%]/ 46,XY,del(1)[80%] mosaicism on blood karyotyping of the father, thus revealing him to be a somaticgonadal mosaic. Normal parental karyotypes do not absolutely exclude the possibility of such mosaicism, as exemplified in two sisters with a chromosome 16 deletion whose parents’ karyotypes on blood were normal (Hoo et al., 1985), and in a case of recurrent, interstitial 1p36 deletion in two sisters Structural Rearrangements •
297
from a gonadal mosaic mother, who did not have any evidence on blood fluorescence in situ hybridization (FISH) analysis of the deletion (Gajecka et al., 2010). A fuller discussion of parental gonadal mosaicism is given on p. 54. A rearrangement arising at a postzygotic mitosis would lead to mosaicism in the child, generally for a normal and for the abnormal cell line. Two major scenarios warrant consideration, in the case of an abnormal child in whom such mosaicism is diagnosed. First, the conceptus is chromosomally normal, and at a subsequent mitosis an abnormality is generated that gives rise to a karyotypically abnormal cell line, along with the 46,N line. The karyotype becomes 46,N/46,(abn). This category will typically have no increased risk for recurrence. Second, from an initially 47,+(abn) conceptus, a postzygotic “correction,” with loss of the abnormal rearranged chromosome, generates a normal cell line, and so the karyotype becomes 46,N/47,+(abn). Here, the recurrence risk will essentially be that of the 47,+abn state. With the exception of marker chromosomes, somatic mosaicism for a structural rearrangement is rarely recognized (Leegte et al., 1998; Zaslav et al., 1999). S I M P LE A N D CO MP LE X DE LE T I ON S
The simple scenario of a “clean-cut” deletion may in some, indeed many instances, be an oversimplification. Davies et al. (2003) restudied a group of 16 deletion patients, and in 3 the supposed deletion proved to be a rearrangement, involving subtelomeric regions. Gunn et al. (2003) studied a child initially karyotyped as 46,XY but whose clinical features suggested an 18q deletion. This was indeed proved, but the deletion seemed rather small given the severity of the phenotype. Using FISH and microarray analysis, they could show that a segment from distal 4q had been inserted into the site of the 18q deletion, giving a partial 4q trisomy along with the partial 18q monosomy. And many examples now exist whereby terminal deletions, apparently simple on light microscopy, prove to be complex rearrangements upon molecular investigation, with submicroscopic duplications, triplications, and inversions, in addition to the deletions (Ballif et al., 2003b, 2007b). Zuffardi et al. (2009) propose that many “deletions,” according to cytogenetic analysis, are actually inverted duplications contiguous with terminal deletions. These result from dicentric chromosomes (which can form either through NAHR or NHEJ), which then undergo a breakage-fusion-bridge 298
•
cycle (Ballif et al., 2003b), with formation of a telomere at the site of rupture. This view is supported by Rowe et al. (2009), who suggest that the most usual mechanism may operate at a premeiotic meiosis, due to NHEJ. COMPL EM EN TA R Y D EL ET I O N / DU PL I CAT I O N
The rare complementary deletion/duplication offers insight into the likely site of generation of this particular rearrangement. If the del/dup should arise at the very first somatic replication following conception, two countertype cell lines will be produced at the two-cell stage, with no normal cell line. In the event that extrafetal tissues can be studied, and still no normal cell line seen, the interpretation of a firstmitosis scenario is strengthened; thus, RodriguezRevenga et al. (2005) could draw such a conclusion from their prenatal diagnostic case of dup(18q)/ del(18q) mosaicism, the chorionic villi showing both karyotypes, although on amniocentesis and fetal blood, only the del(18q) was present. If one of the cell lines is of lesser viability, a child might show the complementary karyotypes at birth, but later in childhood, only one cell line. Morales et al. (2007a) report an example, an abnormal infant who as a newborn had dup(7)(q21.1q31.3)[90]/del(7) (q21.1q31.3)[10] mosaicism, but upon restudy at age 12 and 14 months, only the dup(7) cell line was seen, looking at blood and exfoliated urinary tract epithelial cells. If the del/dup arises at the second (or subsequent) mitosis, there will be a normal cell line as well; and Tharapel et al. (1999) illustrate this circumstance in a child initially identified at amniocentesis, undertaken upon the basis of a choroid plexus cyst and echogenic bowel. In this child, the normal cell line was present in about half of cells, with the remaining cells containing either a deletion for 7p11.2-p13 or a duplication for this segment. I NF LUEN CE O F SEX O F PA R EN T
Certain duplication/deletion rearrangements may have a predilection for happening in the parent of one or other sex. Chromosome 17p11.2 rearrangements are more often of paternal origin, and they may be intrachromosomal or interchromosomal in their generation. The nearby 17p12 region is also more susceptible to rearrangement in the paternal gonad, although in contrast to 17p11.2, paternal duplications/deletions are always interchromosomal, and
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
the uncommon maternal cases are all intrachromosomal (Potocki et al., 2000a). The X chromosome has a particular vulnerability in the male, perhaps because it is largely unpaired at meiosis, and it can refold up and down its length (Giglio et al., 2000). The 1p36 deletion varies in size, and the larger deletions are more often of paternal generation (Gajecka et al., 2007). We may also note that de novo Robertsonian translocations are almost always maternal in origin (Bandyopadhyay et al., 2002), while on the other hand, the great majority of de novo apparently balanced translocations and complex rearrangements arise in the fathers (Grossmann et al., 2010; Thomas et al., 2010). The frequency of paternal deletions and duplications shows a small increased age effect, and with some chromosomes being more susceptible than others (Templado et al., 2011). THE NEED F O R A TELOM E RE
Respecting the requirement that integrity of the telomere be maintained, some mechanisms of terminal deletion need to include a process to restore the telomere (Ballif et al., 2000a; Daniel et al., 2008). If the terminal deletion is interstitial (thus, actually subterminal), then the original telomere simply remains intact. If, however, the telomere is lost in the deletion process, a neo-telomere can be generated (“telomere healing”). If another chromosome is involved in the process, its telomere can be “captured” to fulfill the requirement. Ballif et al. (2003) made particular study of the 1p36 deletion (see later discussion) and showed that all three mechanisms were involved, the commonest being the acquisition of a telomere from another chromosome.
Deletion We have traveled a distance from the earliest days of cytogenetics when the first deletion was published, which was large enough to be seen on a solid-stained “B group chromosome” and associated with cri du chat syndrome (Lejeune et al., 1963). We have, now, a spectrum from large deletions (“classical cytogenetic deletion syndromes”), through microdeletions detectable only since the use of high-resolution banding, to deletions beyond the range of banding
but detected on combined molecular/cytogenetic (FISH) or purely molecular methodology (array comparative genomic hybridization [array-CGH]), and deletions which are so small that only a single gene is removed. Examples of the general karyotypic form of an interstitial deletion, in any chromosome A, are 46,del(A)(p21p23) or 46,del(A)(q12q12). CON T I G UO US G EN E SYN D R O M E
Recollect that loci are arranged in linear order along a chromosome. Often there is no apparent reason for the order: the nonsignificance of the contiguity of two loci has been likened to the unimportance one would attach to “Appalachian Mountains” being next to “apple” in an encyclopedia. Our genome differs from an encyclopedia in that about a third of all the entries relate to one topic: the development and functioning of the brain. Many of the other entries (loci) relate to the control of morphogenesis during embryonic life. If a length of chromosome is deleted, a sequence of adjacent (contiguous) genes will be lost. The phenotype resulting from this can be described as a contiguous gene deletion syndrome (Schmickel, 1986; Tommerup, 1993). In almost any deletion detectable cytogenetically, some of the deleted loci will be brain loci, while others could be for anything, but probably including some morphogenesis loci. Thus, we have the classic clinical picture in deletion syndromes of intellectual deficit of some degree, dysmorphism, and organ malformation. The deletion produces a monosomy—or “haploinsufficiency”1—for the region of the chromosome that has been removed, and loci in this segment are underexpressed. Proof that genetic expression is reduced by 50%, for example, in the case of the 18q–syndrome (see later), was adduced by Wang et al. (1999b) in measuring mRNA from a number of 18q loci. Some of the loci whose haplo-insufficiency contributes to the phenotype in the various deletion syndromes are beginning to be defined, as noted in individual entries in the “Genetic Counseling” section. It seems likely that many such genes will have their untoward outcome not in a simple one-to-one relationship with a single gene product, but rather in a complex layering and interlacing of consequential effects. As yet, however, it is only the simple case
1 This word can be, and is, used in the context of a single locus. It is usually applied in cytogenetics to the more recently delineated “microdeletion” syndromes, but it can in principle refer to the classical syndromes with larger deletions.
Structural Rearrangements •
299
that we can begin to understand: such as, for example, the brain white matter abnormality of the 18q– syndrome just mentioned, that is presumably a direct consequence of the loss of a structural myelin gene on 18q. As array-based CGH may define deletions/duplications of quite small extent, only a few genes may be located within the particular segment, and the counselor blessed with a scientific curiosity has the opportunity to check which genes these are, and perhaps to make an informed speculation (which some parents might find helpful) as to which of these might have contributed to a child’s phenotype (and see section on “Gene Discovery” later). If two or more mendelian disorders coexist in the one person, a contiguous gene deletion is a strong possibility. In order to prove the point, molecular methodology can be brought to bear; or a direct chromosomal test using FISH offers an immediate visual demonstration of the deletion. We have, for example, seen a young woman presenting with a history of recurrent bacterial infections since childhood, and night blindness and diminishing peripheral vision since teenage, leading to diagnoses of chronic granulomatous disease and retinitis pigmentosa (Coman et al., 2010b). The X-linked forms of these conditions being very closely linked, a contiguous gene deletion suggested itself, and a FISH probe targeted to a DNA sequence between the two loci was generated. Its nonhybridization to one X chromosome essentially confirmed the supposition of a deletion. Furthermore, this led the way to another diagnosis, that of a partial protein intolerance, due to deletion of the OTC (ornithine transcarbamylase) gene, which lies in the same Xp region. An infrequent mechanism for abnormality is that the deletion may “unmask heterozygosity” for an autosomal recessive disorder, and there are a handful of examples of this having been recognized (Coman and Gardner, 2007). Flipsen-ten Berg et al. (2007) reported an infant with Wolf-Hirschhorn syndrome (WHS) due to a deletion on one chromosome 4p, and who then went on to develop, over and above the WHS, signs of Wolfram syndrome (diabetes mellitus, diabetes insipidus, deafness, optic atrophy). The WFS1 locus for Wolfram syndrome is on chromosome 4 short arm, which led these workers to examine the gene on the “normal” chromosome 4. A point mutation in WFS1 was discovered. The 4p deletion on the 300
•
other chromosome allowed this mutation to be “exposed”; and the child, being essentially hemizygous, got the syndrome. Similarly, a child with a 22q13 deletion (Phelan-McDermid syndrome), and having an ARSA mutation on the other chromosome 22, would develop the fatal recessive brain disease, metachromatic leukodystrophy (Bisgaard et al., 2009). Subtelomeric Deletions. The subtelomeric region is typically gene-rich and may be more prone to deletion. A very subtle deletion may submit only to FISH and molecular methodology, and somewhere in the vicinity of 2%–10% of patients with “unclassified multiple congenital anomalies/mental retardation” (MCA/MR) may prove to have a subtelomeric deletion. Microarray is the more powerful of the two methodologies, in terms of the yield of abnormalities (Ballif et al., 2007c). A less affected parent might have the same deletion, and thus parental studies may be warranted (notwithstanding a note referring to “parents normal” on the test requisition form). Some apparent subtelomeric “deletions” may actually be due to normal polymorphism (Chapter 16), and this is another reason for parental chromosomes to be studied, along with careful clinical assessments, carefully interpreted (Ballif et al., 2000a). Microdeletions and Mendelian Disorders. Several examples exist of mendelian syndromes, the majority of cases representing classic single-gene mutations, but a few being the consequence of haplo-insufficiency due to microdeletion that deletes the entire gene, and often neighboring genes as well. Rubenstein-Taybi syndrome (see later) was one of the earliest to exemplify this scenario (the gene CBP); and Charcot-Marie-Tooth disease type 1A and hereditary pressure-sensitive palsy provide an instance of both duplications and deletions with respect to a single gene (PMP22) (see later), and indeed this genomic mechanism being more frequent than classical mutation at this locus. Other cases, most of which are also noted later, include: Pitt-Hopkins syndrome (the gene TCF4), Sotos syndrome (NSD1), Alagille syndrome (JAG1), SaethreChotzen syndrome (TWIST), Greig syndrome (GLI3), one type of holoprosencephaly (SHH), CHARGE syndrome (CDH7), one type of lissencephaly (LIS1), and X-linked ichthyosis (STS). If neighboring genes are also deleted, this may contribute to a wider phenotype; or these other genes
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
might not be dosage sensitive, and the clinical picture is essentially the same as in the mendelian case. A number of these loci were originally discovered due to patients having deletions of the regions concerned. Gene Discovery. Taking a broader view, deletions can point the way to discovery of novel genes coding for particular organs and tissues. A deletion that, from clinical observations, has a particular clinical association with clefting, may remove, say, contiguous genes w, x, y, and z. It could reasonably be imagined that haplo-insufficiency of one of these genes (perhaps gene x, if its product had a theoretical role in cell-cell interaction) could contribute to the cause of cleft lip; and then it could further be assumed that the normal role of gene x is in contributing to the process of lip formation during early
embryogenesis. Brewer et al. (1998) have reviewed some hundreds of small deletions listed in the Oxford Cytogenetic Database, correlating the malformations with which these deletions have been associated. Patterns have emerged: some deletions seem particularly likely to lead to a heart defect, while others may be prone to cause clefting. A larger scale map, addressing only the distal long arm of chromosome 13, is shown in Fig. 19–2. This cartographic approach, broad-brush in the 1990s, we may expect to become progressively more finely drawn as this new century advances. Researchers hoping to find genes directing development of the heart, or genes controlling lip formation, could focus their searches in these chromosomal regions. Similarly, nephrogenesis genes and neurogenesis genes may come to light in the analysis of deletions associated with renal maldevelopment
Chromosome 13q Mb 13q12.11
13q13.3
13q14.2 13q21.1 13q21.2 13q21.31 13q21.32 13q21.33 13q22.1 13q22.2 13q22.3 13q31.1 13q31.2 13q31.3 13q32.1 13q32.2 13q32.3 13q33.1 13q33.2 13q33.3 13q34
54 61
Micrognathia
83 89 99 103 105 108 114
Short stature
DWM
CP/CL
DWM+ACC+NTDs Prominent columella Microcephaly
Cortical heterotopia
Thumb a-/ hypoplasia an-/ microphthalmia Thumb a-/ hypoplasia
Distal limb anomalies association
Lung hypoplasia Anal atresia + penoscrotal inversion
FIGURE 19–2 A deletion-phenotype map for deletions of distal 13 long arm, from 13q21.1-qter. The application of microarray comparative genome hybridization (CGH) allowed considerable precision in the delineation of the deleted segments. ACC, agenesis of corpus callosum; CP/CL, cleft palate with or without cleft lip; DWM, Dandy-Walker malformation; NTDs, neural tube defects. (From M. Kirchhoff et al. 2009, Phenotype and 244k array-CGH characterization of chromosome 13q deletions: An update of the phenotypic map of 13q21.1-qter, American Journal of Medical Genetics 149A: 894–905. Courtesy M. Stefanova, and reproduced with the permission of Wiley-Liss.) Structural Rearrangements •
301
and epilepsy, respectively (Singh et al., 2002a; Amor et al., 2003). Different syndromes with a phenotypic trait in common can clarify the nature of the genetic heterogeneity. Thus, multiple exostoses are a component of the Langer-Giedion syndrome, due to deletion at 8q24, and the Potocki-Shaffer syndrome, due to 11p11.2 deletion, consequent upon haploinsufficiency of different bone growth genes at these two segments. Rare Complexity. A rare (or rarely recognized) complexity is a change in size, from parent to child, of a subtelomeric deletion. Faravelli et al. (2007) observed a mother having a 1.5 Mb 4p deletion, which expanded in size to produce a 2.8 Mb deletion in her child with typical Wolf-Hirschhorn syndrome; in retrospect, the mother had subtle signs to suggest a forme fruste. Similarly, South et al. (2008a) describe a normal mother with a de novo 18q subtelomeric deletion of 0.4 Mb, which increased in size nearly 10-fold, to 3.7 Mb, in her abnormal daughter, who had presented with a clinical picture consistent with 18q deletion syndrome. This parent-to-child “expansion” is likely due to a recombination mechanism.
Duplication Duplicated segments may arise from within the same chromatid, from the sister chromatid, from the same arm, from the other arm, or from a different chromosome, through the similar mechanisms as described earlier. For a number of segments, both deletions and duplications have been recognized, and these cases are listed in Table 19–1. As with the deletion, the association of different duplicated segments with particular phenotypes offers an insight into which regions of the genome may harbor specific critical genes (Brewer et al., 1999). In general, identification of the phenotype-causing genes from duplications is more difficult than deletions, with partial trisomy typically having a lesser phenotypic consequence than partial monosomy. In some (by no means all) cases, the duplication (or sometimes triplication) of a locus manifests as the “opposite” phenotype as that found in the deletion. Thus, triplication of the MMP23 gene on 1p36 results in craniosynostosis, while the deletion is associated with a large, late-closing anterior fontanel (Gajecka et al., 2005). Deletion of the P gene in the Prader-Willi/ Angelman region causes lighter complexion (and when mutated causes oculocutaneous albinism type 2); an 302
•
Table 19–1. Some Chromosomal Syndromes for Which Both Deletion and Duplication of the Same Microsegment Have Been Described del
dup
dup dup dup* dup dup dup dup dup dup dup dup dup dup
15q13.3 15q24 16p11.2 16p13.11 16p13.3 17p11.2
del del Albright-like del Wolf-Hirschhorn Cri du chat Sotos Williams del Potocki-Shaffer WAGR Congenital Rett Prader-Willi, Angelman del* del del del Rubinstein-Taybi Smith-Magenis
17p12 17p13 17q12 17q21.31 20p12 22q11 22q11.21-q11.23
HPSN Miller-Dieker del del Alagille diGeorge del
1q21.1 1q44 2q37 3q29 4p16.3 5p13 5q35 7q11.2 10q22q23 11p11.2 11p13 14q12 15q11-q13
dup* dup** dup dup* dup PotockiLupski CMT dup dup dup dup dup dup*
Notes: Some syndromes have an eponymous nomenclature; most are noted simply on the basis of del or dup of the involved segment. Of these, Charcot-MarieTooth neuropathy (late nineteenth century), Angelman, cri du chat, diGeorge, Prader-Willi, Sotos, Rubinstein-Taybi and Williams syndromes, and pressure-sensitive neuropathy (mid-twentieth century), and Alagille (later twentieth century) are the only ones to have been described well in advance of the recognition of their chromosomal/genetic basis (“phenotype-first”). *Phenotype, in some at least, may have been considered to lie within a normal range. **Single case, inherited from normal father; causal link uncertain (Kiholm Lund et al., 2008). CMT, Charcot-Marie-Tooth neuropathy; HPSN, hereditary pressure-sensitive neuropathy. Source: All entries from published reports, other than for dup 11p11.2, which is a personal communication from J. M. Gastier-Foster (2009).
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
increased copy number of this gene leads to hyperpigmentation (Akahoshi, 2004). Shinawi et al. (2010) refer to these opposite scenarios as “sister genomic disorders” (and see later, dup 16p11.2). Direct and Inverted Intrachromosomal (Tandem) Duplication. The duplication comprises chromatin of the same chromosome, the original and the duplicated segments being ordered in tandem fashion. If the linear orientation of a chromosome A is maintained, the rearrangement is a direct duplication, 46,dir dup(A); if it is reversed, it is an inverted duplication, 46,inv dup(A). As noted earlier, many apparent inverted duplications may in fact exist along with a terminal deletion; the causative mechanism may be either to NAHR or NHEJ, and most often occurring at a premeiotic mitosis (Rowe et al., 2009; Zuffardi et al., 2009). The inv dup/del 8p is a well-recognized recurring example (Kotzot et al., 2000b; Kondoh et al., 2003). An alternative explanation, based on an analysis of many cases of complex rearrangements of 1p36, is that successive breakage-fusion-bridge cycles result in deletion/duplication chromosomes, particularly near to the telomeres (Ballif et al., 2003b). Faivre et al. (2000b) propose a postzygotic sister chromatid exchange in a child with a condition resembling Sotos syndrome and having mosaicism for a dup(20) (p11.2-p12.1) in 23% of cells. On checking DNA markers that mapped to the region of the duplication in 20p, each parent had contributed just one allele. The parsimonious explanation is that a single event of unequal sister chromatid exchange happened at a mitosis, in an initially normal conceptus, generating the dup(20p) line, with the complementary deletion being lost (Fig. 19–3). One rather subtle intrachromosomal duplication, and possibly triplication, needs to be distinguished from a normal variant. This is the “dup(8) (p23.1),” listed as a euchromatic variant in Chapter 16 (p. 264). On classical cytogenetics, the two forms are indistinguishable. But a replication of certain sequences within this region, different from those underlying the normal variant, can produce a phenotype due to a dosage effect of these segments (Barber et al., 2005). The phenotype may include minor cognitive impairment, mild facial dysmorphism, and structural heart defect, the latter possibly due to perturbation of the GATA4 locus. Such cases typically arise de novo, and this is a clinical pointer enabling recognition of this pathogenic dup/trp.
FIGURE 19–3 A mechanism whereby a mosaic duplication and/or deletion could be produced. An unequal sister chromatid exchange at a postzygotic cell cycle generates a chromosome with a duplication (dup) in one chromatid and a deletion (del) in the other. At the next mitosis, the two chromatids segregate to the daughter cells, giving rise to a dup cell line and a del cell line. The del line may be lost, since partial monosomies are generally less survivable than partial trisomies, in which case only the cells of the dup lineage would exist alongside the normal cells. (After Faivre et al., 2000b.)
Additional Material from Another Chromosome. In other rearrangements, the duplicated material has come from another chromosome. An example of the karyotype nomenclature, to be used before the nature of the additional material has been established, is 46,add(1)(q36). Pairing between nonhomologs, followed by crossing-over (due to NAHR or NHEJ), produces reciprocal products that are two derivative chromosomes. In a “single-segment” exchange, one of these will have a duplication, and the other a deletion. If this occurred during meiosis, and if segregation were then asymmetric, gametes with a duplication or with a deficiency would be produced. Other scenarios, with more complex Structural Rearrangements •
303
mechanisms, may be imagined. Coles et al. (1992), for example, studied a child with Wolf-Hirschhorn syndrome who had two separate de novo rearrangements of the X chromosome with a chromosome 4 and the Y, respectively, and they propose that simultaneous or sequential crossovers happened in a meiotic “octad” of four synapsing chromosomes. If a de novo unbalanced rearrangement could be shown to have its component parts originating from a maternal and a paternal chromosome, the fact of its postzygotic origin would be thereby demonstrated. Sarri et al. (1997) offer an example of this scenario in a malformed child with 46,X,der(X), t(X;17)(q27;q22) whose der(X) originated from the paternal X and the maternal 17 chromosomes. Eggermann et al. (1997) report a similar case, an abnormal child with a de novo der(18)t(13;18) (q14.3;q23). The chromosome 18 component of the translocation came from the paternal chromosome 18, and the chromosome 13 component from the maternal chromosome 13. In this type of biparental rearrangement, even the very small theoretical risk otherwise associated with parental gonadal mosaicism could confidently be excluded. Similarly, mosaicism in the presence of a normal cell line would typically allow the presumption of a mitotic origin. Zaslav et al. (1999) report a child with a severe brain malformation who had the karyotype 46,XX,der(4)t(4;15)(q35;q22)/46,XX. They propose that the chromosome constitution at conception was 46,XX. At an early cell division, a reciprocal exchange occurred between chromatids of chromosomes 4 and 15. Then, at anaphase, there was an unfortunate segregation. The newly generated der(4) passed to one daughter cell, along with the normal chromosome 15; and, vice versa, the der(15) and the chromosome 4 passed to the other. The former produced a cell line with a del(4)/ dup(15) imbalance, and the presence of this cell line in the developing nervous system presumably caused the brain maldevelopment. The other cell line was not seen (on a peripheral blood karyotype), and it may have been selected against. (If the segregation of the chromosomes at that crucial mitosis had been balanced, then the child would likely have been a phenotypically normal mosaicism balanced translocation carrier.) Reddy and Mak (2001) could demonstrate mosaicism in both blood (conventional karyotyping) and on buccal mucosal cells
(FISH) in two patients with additional material from another chromosome. For example, one patient had mosaicism for an add(5), the additional material coming from 3p26-pter, in 32% of lymphocytes. Using a 3p-subtelomere probe, a very similar level of mosaicism (40%) was shown in buccal epithelial cells. Triplication. A very few cases are known, at the level of classical cytogenetic analysis, of a segment of chromosome replicating twice over, being in threefold amount on that homolog. The segment is thus present, in total, in four-fold dose. Triplications observed on classic cytogenetics are reported for chromosomes 2, 4, 5, 7, 9, 10, 12, 13, 15, and 16; in the triplication 12p case, the phenotypically normal mother was a low-level (12%) mosaicism for the same rearrangement (Eckel et al., 2006). On microarray, we have seen several more cases of triplication, and it seems likely that this “rare” complexity may be less rare than initially supposed. Familial transmission is recorded: the 46,XX,trp(4)(q32.1q32.2) mother in Wang et al. (2009b) had three sons with the same imbalance, 46,XY,trp(4)(q32.1q32.2), this being clearly visible on classical karyotyping.
Supernumerary Marker Chromosome Many different supernumerary marker chromosomes (SMCs) exist, in the general karyotype 47,+mar (mar for “marker”), or 47,+SMC; they are also known as extra structurally abnormal chromosomes (ESACs), and accessory chromosomes.2 Small ones are abbreviated as sSMCs, and very small, minute ones as minSMCs. The 47,+SMC individual has a duplication (partial trisomy), or in some cases a triplication (partial tetrasomy) of the material comprising the SMC. The birth prevalence is in the range 2–7 per 10,000. Blennow et al. (1995) record a large Scandinavian experience: of 50 SMCs, almost half were idic(15), six were small rings deriving from various autosomes, six were isochromosomes of 18p or 12p, and most of the remainder were harmless SMCs derived from acrocentric chromosomes. A particular category is the SMC that lacks α-satellite DNA (a component of the normal centromere) but which possesses a “neo-centromere,” a point of considerable theoretical interest and possibly practical significance (see later section on “Rare Complexities”).
2 For the record, the first such case, from Australia, was reported as “incomplete trisomy” (Ilberry et al., 1961).
304
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
With increasing sophistication of staining techniques, not to mention the advent of array-CGH,3 the expression “SMC” is to be seen as a temporary designation, awaiting the full delineation of whatever partial trisomy it may be. Readers who delight in bold colors should refer to Reichenbach et al. (1999). These authors describe a child with 47,+mar who, upon multicolor banding, could be seen as having the karyotype 47,+del(5)(q11). Guanciali-Franchi et al. (2004) used spectral karyotyping to identify a collection of 14 previously cryptic SMCs. By contrast, an absence of color was, in a sense, even more dramatic in a mildly abnormal child studied by Mackie Ogilvie et al. (2001): he had a C-band negative SMC that failed to show hybridization with any whole chromosome paint and could not be identified using a range of other cytogenetic methods (it also lacked a conventional centromere). The origin of this SMC was thus quite baffling. Liehr (2008) reviews the subject at length, and he has a Web site devoted to the SMC: http://www. med.uni-jena.de/fish/sSMC/00START.htm
Isochromosomes The classical isochromosome is a “mirror-image” chromosome, with two identical arms either side of the centromere; a more complicated rearrangement is the isodicentric (or inverted duplication) chromosome. When present as a supernumerary chromosome, the classic isochromosome imposes a tetrasomic state for the chromosomal arm concerned. Recorded isochromosomes include i(5p), i(8p), i(9p), i(10p), i(12p) (Pallister-Killian syndrome),4 i(18p), i(18q), i(20p), i(22q), and i(Xq) (not including the Robertsonian acrocentric isochromosomes, which are dealt with in Chapter 7, and the i(21q) in Chapter 18). Isochromosomes can arise by a variety of mechanisms and at diverse times and places. The most simple, and classical, mode of isochromosome formation is a misdivision at the centromere, also known as “centric fission” (Rivera and Cantú, 1986). It can be thought of as a horizontal rather than a vertical division (Fig. 19–4). This gives rise, in a chromosome A, to short arm and long arm isochromosomes, an i(Ap) and an i(Aq).5 More complex scenarios can
be devised, such as the U-type exchange, with the chromatid of one arm of a chromosome “looping around” to join with its fellow. Whatever the mechanism, the process could occur: in a premeiotic gametocyte (which would reflect a gonadal mosaicism); during one or other meiotic division; in the zygote; or at an early or a later postzygotic division, in an initially normal or an initially trisomic conceptus. The isodicentric chromosome, when present as a supernumerary chromosome, 47, +iso, typically arises following aberrant homologous recombination in meiosis. Subsequent mitotic loss of an isochromosome in one cell lineage can bring about a mosaic state, potentially having a less severe functional imbalance, which is not necessarily a fortunate event, since an otherwise early in utero lethal imbalance could convert to a survivable but profoundly abnormal phenotypic state. An interesting sequence of events is proposed in de Ravel et al. (2004), concerning the case of an abnormal fetus karyotyping 47,XX,+12/47,XX,+i(12)(p10) post termination. Initially, a disomic 12 egg met a monosomic 12 sperm, producing a trisomy 12 zygote. From one cell, a 47,+i(12p) mosaic line was created, due to rearrangement of the paternal chromosome 12 (and a normal cell line was also produced, seen only in short-term CVS culture). A similar scenario, with the postzygotic separation of a normal and an isochromosomal cell line, may have been the basis of karyotypically discordant, presumed monozygous twin fetuses, one with 47,XY,+i(5p), and the other 46,XY (Grams et al., 2011). In contrast, the isochromosome replacing a normal homolog, 46,iso, is almost always postzygotic in origin, from an initially normal conceptus, as for example Riegel et al. (2006) showed in two babies with Down syndrome and Patau syndrome. A normal karyotype had been seen at CVS, but the babies subsequently born karyotyped 46,i(21q) and 46,i(13q), respectively. The very rare circumstance of recurrence of an isochromosome in siblings presumably reflects a premeiotic generation of the abnormality in a parental gonad (Krüger et al., 1987). The error in this circumstance could have occurred at mitosis in the parent, some distance along in their embryonic development, with only a small fraction of the body
3 But one should be aware that low-level mosaicism for a SMC could be missed by array-CGH (Ballif et al., 2006, 2007b). 4 Magenis et al. (1999) record the historical point that Pallister-Killian syndrome was first identified serendipitously, when fibroblasts taken for archival purposes were subject to routine cytogenetic analysis. 5 The band p10 (or q10) is sometimes noted, as for example, in 47,XX,+i(12)(p10), indicating that each arm of the isochromosome comprises the entire p (or q) arm.
Structural Rearrangements •
305
(minimally, a part of one gonad) involved. Or the error could have been present at the parent’s conception, with a postzygotic loss of the supernumerary isochromosome in one cell line which becomes 46,N, and favorable distribution of this line in the soma, but not gonad, thereafter. Boyle et al. (2001) propose such an evolution in a family in which a mother, 46,XX on blood karyotype, had had two children (half-sisters) with nonmosaic 47,XX,i(18p). She herself, therefore, must have been a gonadal mosaic. As these workers show, it is likely that she had been 47,XX,i(18p) at her own conception, due either to a meiosis II error or a premeiotic mitotic error in her own mother’s oögenesis. In her postzygotic development, the i(18p) line was lost in most tissue, but not in gonad. And surely, one would have thought, gonadal mosaicism was the reason for the father in Williams et al. (2001) having had one child with i(18p) and another with i(18q). A centromere misdivision in an early gametic stem cell mitosis was the obvious explanation. But on sperm analysis, over 1000 cells counted had just a single 18p and a single 18q signal. Examples like these oblige some caution in counseling, although it is true that such cases are very rare.
FIGURE 19–4 Outline of the classical theoretical mechanism to produce an isochromosome, by “horizontal misdivision” at the centromere. In this example, the site of its generation is at the first division of the zygote, considering the case in point of the i(21q) form of Down syndrome. The normal zygote has two chromosome 21 homologs (maternal crosshatched, paternal open). At the first mitosis, the maternal chromosome 21 divides appropriately at the centromere to give two normal daughter chromatids, but the paternal chromosome 21 misdivides. One product of the misdivision is an i(21q), and the cell resulting is trisomic for 21q (lower left). The other product is an i(21p), and this cell essentially has a 21 monosomy; its lineage does not survive. Thus, the child has a nonmosaic i(21q), and a typical DS phenotype. If the abnormal cell division occurs at a later mitosis (the second or subsequent), a mosaic 46,i(21q)/46,N karyotype could result.
Isodicentric 15. A supernumerary marker chromosome that warrants special attention is the bisatellited dicentric marker, “idic(15),” also known as inverted duplication 15, inv dup(15), or pseudodicentric 15 (Battaglia, 2008; Wang et al., 2008). The usual mechanism of formation is a translocation event between homologs during maternal meiosis, due to apposition of low copy repeats that are located within 15q11-q14. After formation, there is a nondisjunction event and centromere inactivation. Depending on the sites of recombination, there are several types of inv dup(15), classified according to the amount of euchromatic material present. Generally, they are bisatellited and pseudodicentric (Wandstrat and Schwartz, 2000). The important clinical question is this: Is the Prader-Willi and Angelman critical region within 15q12–13 (PWSCR/ ASCR) included in the chromosome? Probes for D15S10 and SNRPN targeting the PWSCR/ASCR are useful in assessing the amount of material (Eggermann et al., 2002). The markers can be divided into three groups: (1) Very small chromosomes with so little chromatin between the centromeres that the appearance is monocentric, and FISH-negative for the 15q12
306
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
imprinted region with respect to the loci D15S10/ SNRPN. These marker chromosomes are assumed not to be causally associated with an abnormal phenotype, and many times they are inherited from a clinically normal parent. Possibly, there may be an association with infertility in the male (Oracova et al., 2009). (2) Medium-size chromosomes with two distinct centromeres with visible intervening chromatin. Because it is difficult to distinguish these from the next group, FISH with D15S10/SNRPN should be performed to be sure that the PWSCR/ASCR segment is not present. An inv dup(15) lacking this segment is typically not associated with an abnormal phenotype. (3) Medium and larger (greater in size than a G-group chromosome) chromosomes, and which stain positively for D15S10/SNRPN, and which thus include the PWSCR/ASCR region. The presence in trisomic or tetrasomic dosage6 of the PWSCR/ ASCR segment—and in particular when of maternal origin—correlates with abnormality, and the phenotype encompasses pervasive developmental disorder with autism, epilepsy, and minor physical defects (Battaglia, 2008). Thus, a reasonable approach to sorting out the various types of inv dup(15q) marker chromosomes, after identification on G-banded analysis, includes FISH for the 15q12 imprinted region (D15S10/ SNRPN) on metaphase chromosomes. If the region is present, interphase FISH can be used to examine whether there is one or two copies. If it is unclear whether the marker is of 15 origin, one might choose to go directly to microarray analysis; indeed, the initial recognition may have been based upon this methodology. This would also enable an assessment of the amount of euchromatin involved, albeit that there is not necessarily a strong correlation between the copy number quantum of some imprinted loci and the clinical phenotype (Hogart et al., 2009). Another caveat is that microarray analysis may not detect low-level mosaicism. Isodicentric 22: Cat-Eye Syndrome. One of the better known SMCs is the inv dup(22)(pter-q11.2) of the “cat-eye syndrome,” and Mears et al. (1994) and Rosias et al. (2001) provide reviews. Two loci CECR1 and CECR2 may be critical dosage-sensitive
genes (McDermid and Morrow, 2002). The region that is duplicated can vary, in some cases extending to include the region of the dup22q11 syndrome (Yobb et al., 2005). The chromosome is not necessarily symmetrical (not truly “iso”), depending upon the actual sites of homologous recombination within 22q11 that led to the rearrangement, and the euchromatic region may be present in trisomic or tetrasomic state. Most cases arise de novo, but familial transmission is recorded, including, remarkably, familial mosaicism (Urioste et al., 1994b). The phenotype appears not to correlate well with the size of the chromosome and indeed the person may show no signs of the syndrome (Crolla et al., 1997); the characteristic heart defect is total anomalous pulmonary venous return. Bergman and Blennow (2000) describe the unique case of a phenotypically normal man with inv dup(22) mosaicism, who also had a 22q11 deletion and a ring 22; it is plausible that the three abnormal chromosomes arose from related recombinations. RA R E CO M PL EXI T I ES
Supernumerary Marker Chromosome with Neocentromere. Most neocentromere-SMCs exist as an inverted duplication, with the neocentromere forming on one of the two otherwise identical “arms” (rather than at the inv dup breakpoint). The two “arms” add up to a tetrasomic state (Voullaire et al., 2001; Mascarenhas et al., 2008). These neocentromeres lack α-satellite DNA and its centromere binding protein (CENPB) (hence the alternative name “analphoid centromere”). In other respects, neocentromeres function similarly to normal centromeres as evidenced by mitotic stability (in many, but not all cases) and by the binding of other known centromere proteins, such as CENPA, CENPC, and CENPE (Saffery et al., 2000; Voullaire et al., 2001). Most chromosomes are now represented in the list of those in which a neocentromere has been identified. Neocentromerization can be considered as a process based upon an epigenetic mechanism: the DNA in this region is the same as in the normal chromosome, but it has now been influenced to take upon itself a new identity and function (Amor and Choo, 2002). More complexities, and including the suggestion of a role for the neocentromere in
6 The presence of two supernumerary idic(15) chromosomes, or one marker with four copies of the PWSCR/ASCR, offers the rare opportunity to use the word “ hexasomy” (Qumsiyeh et al., 2003; Hoppman-Chaney et al., 2010). The same principles apply in terms of presence/absence of SNRPN, with the abnormal phenotype being aggravated in the hexasomic state.
Structural Rearrangements •
307
evolution, are addressed in Amor et al. (2004); an example of a neocentromeric chromosome providing balance for a corresponding deletion is noted later (del 11p11.2). Centromere-Telomere Fusion. Centromeretelomere fusion is a rare mechanism to form an isochromosome. So far, this has been observed with an isochromosome for the short arm, and the long arm of that chromosome being attached to the telomere of another. Rivera et al. (1999b), for example, describe an iso(12p), in which the 12q element was translocated to 8pter. They could show FISH signals for both centromere and telomere probes at the 8pter/12cen union point in the translocation chromosome. The probable mechanism is a postzygotic centric fission of the 12, with the 12q element combining through a centromere-telomere fusion, and with the 12p element doubling up to produce the isochromosome. The end result was trisomy for 12p. Besides 12p, this scenario has been observed in isochromosomes for 4p, 5p, 7p, 9p, and 10p. All cases have been de novo. Complementary Isochromosomes. This very rare circumstance is dealt with in Chapter 8.
Apparently Balanced But Actually Unbalanced Rearrangement If, on classical cytogenetics, an apparently balanced de novo translocation, inversion, or insertion is discovered in a patient with a “chromosomal” phenotype of dysmorphism/anatomical abnormality and developmental delay (young child) or cognitive compromise (older child, adult), it is logical, and sometimes correct, to assume a causal link (Tonk et al., 2003). Further cytogenetic analysis may reveal that there is, in fact, an actual imbalance. With the advent of array-CGH, a clearer light, and often greatly clearer, can be cast upon these cases (Gajecka et al., 2006; De Gregori et al., 2007; Baptista et al., 2008; Sismani et al., 2008). Loss or gain of DNA at the site(s) of rearrangement provides strong supporting evidence in favor of a pathogenic effect, and particularly so if specific genes can be implicated. Conversely, a normal quantitative result would point to the rearrangement being truly balanced, although the possibility of disruption of a gene at the actual site of a breakpoint could yet be the explanation for an abnormal clinical picture (Cacciagli et al., 2010). Occasionally, the result is 308
•
surprising: there may be no DNA imbalance at the site of breakpoints, but a deletion and/or duplication is seen at some other place in the genome. Here, the rearrangement can presumably be exonerated, as a coincidental event; the real cause lies elsewhere. Note that if the rearrangement that disrupts the gene is a “clean” break, with little or no loss of DNA, then a microarray would not detect it. Position effect is another mechanism whereby a “balanced” rearrangement can lead to phenotypic abnormality. The SOX9 gene on chromosome 17 at band q25.1, the basis of campomelic syndrome, provides an example with respect both to a translocation and an inversion. The de novo translocation t(5;17)(q15;q25.1) in Figure 19–5 was seen in a child with this syndrome, as was the de novo paracentric inversion inv(17)(q24.3q25.1) reported in Maraia et al. (1991). Other such examples are mentioned later (see del 7q21.3, del 11p11.3). This type of rearrangement would probably not be detectable on microarray.
GENETIC COUNSELING Deletions and Duplications In most children with deletions or duplications, the parents type as 46,XX and 46,XY on routine blood analysis, and the defect is “de novo.” The risk for recurrence is very small, but it is not nonexistent. An example is noted later under “del 5p”; recurrence of a typical Prader-Willi deletion in brothers is recorded in Fernández-Novoa et al. (2001); and a del(9) (q12q21.32) has been seen in siblings (M. D. Pertile, personal communication, 2010). Röthlisberger and Kotzot (2007) undertook a review and were surprised at how few actual cases had been published. The rare recurrences are likely due to an occult parental mosaicism, which the routine blood chromosome study could not detect. The abnormal line in the parent may be gonadal (confined to gametic tissue) or somatic-gonadal (some somatic tissues involved as well—but not, apparently, blood). The observation of rarity of recurrence allows us to propose the empiric advice that, in the individual case, recurrence is most unlikely. A figure that is appropriate in this setting might be “less than ½%”; the counselor should note the converse “greater than 99½%” for a child without the chromosome defect. Even this may be an overstatement: Röthlisberger and Kotzot (2007) consider that “less than 0.3%” may be closer to the mark. Some couples may find
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
FIGURE 19–5 An apparently balanced translocation causing the syndrome of campomelic dysplasia (which includes skeletal, genital, and brain defects). One breakpoint is at 17q25.1, on or close to the SOX9 locus (shown as dot on the cartoon karyotype), where the basis of the syndrome lies. One possibility is that the gene is disrupted. Or, an influence of adjacent chromosome 5 chromatin (“position effect”) leads to inactivation of the SOX9 gene on the der(17), the functional SOX9 haplo-insufficiency then being responsible for the phenotype. (Case of R Savarirayan; Savarirayan and Bankier, 1998.)
“greater than 99.7%” sufficiently encouraging that they would not request prenatal diagnosis in a subsequent pregnancy. Some, however, might; and one could sympathize with this request. If testing is to be based upon classical cytogenetics alone, care must be exercised in offering prenatal diagnosis of small deletions/duplications seen only on high-resolution lymphocyte chromosomes. The technical ability to demonstrate such small changes in amniotic fluid or chorionic villus cells may be limited. In those deletions/duplications where a parent is shown to carry a balanced rearrangement, a substantial recurrence risk is probable, and the appropriate chapter should be consulted. Rarely, the same deletion/ duplication might be seen, somewhat unexpectedly, in a parent, an observation that underpins the advice that parental karyotyping does need to be undertaken (Sparkes et al., 2009). Extremely rarely, an inverted duplication may, in fact, be due to recombination within a parental paracentric inversion (p. 181). Paracentric inversions can be difficult to detect, and a careful and directed search may be appropriate. With the increasing use of powerful molecular cytogenetic methodologies, and with particular reference to array-CGH, many more cases of chromosomal imbalance are being recognized (Shaffer and Bejjani, 2006; Shaffer et al., 2007a; Bejjani and Shaffer, 2008; Slavotinek, 2008). Figure 19–6 lists recorded cases on the European Cytogeneticists Association database (see later). Documentation of such cases is of much value to the clinician, but the sheer volume of new discoveries does represent a challenge. One organ, the European Journal of Medical Genetics, is devoting an occasional section headed
“Chromosomal Imbalance Letters,” and this is likely to prove a useful repository, and likewise DECIPHER is a valuable source (see next section). Internet Resources. A number of databases have been developed, accessible on the Internet, and these are a most valuable resource. We may list the following: • The European Cytogeneticists Association, building substantially upon the work of the group of Professor Albert Schinzel in Zürich, has established a register of unbalanced chromosome aberrations (ECARUCA), which can be accessed—and contributed to—at http://www. ecaruca.net (Feenstra et al., 2006). A particular chromosomal abnormality can be searched for, according to its standard cytogenetic description, or by array-CGH criteria, at the level of the nucleotides encompassing a deletion or duplication. ECARUCA publishes from time to time their totals of deletions and duplications per chromosome, and the July 2009 listing is shown in Figure 19–6. • The UK Chromosome Abnormality Database (UKCAD) provides data from all the UK Regional Cytogenetics Centers and is accessed at http://www.ukcad.org.uk/cocoon/ukcad/ • DECIPHER (Database of Chromosomal Imbalance and Phenotype in Humans using Ensembl Resources) is a repository of phenotypes for chromosome alterations identified by microarrays, and it can be accessed at http://decipher.sanger.ac.uk/ Structural Rearrangements •
309
Deletion Duplication
Deletion Duplication
1 137 82
2 139 101
3 116 113
4 280 91
5 116 75
6 130 70
7 184 85
8 163 199
9 158 161
10 196 102
11 192 132
12 59 69
13
14
15
16
17
18
19
20
21
22
Y
X
152
70
189
49
149
237
5
45
74
283
10
62
73
63
87
88
66
96
21
56
43
139
9
13
FIGURE 19–6 The listing of the numbers of deletions and duplications observed per individual chromosome, from the database of the European Cytogeneticists Association Register of Unbalanced Chromosome Aberrations (ECARUCA), as of July 2009 (http://www.ecaruca.net). (Reproduced with the permission of ECARUCA.)
• Signature Genomic Laboratories have developed an in-house database, “Genoglyphix,” of microarray-based chromosome diagnoses. This is open to the referring clinicians, laboratory diagnosticians, and genetic counselors, who can generate their own databases through this system. The Web site address is http://www. signaturegenomics.com/genoglyphix.html D E L E T IO N S
Brief sketches of the major deletion syndromes follow, as well as some less well-known ones, in numerical order of chromosomes, and numerical order of p and q segments. Some gain inclusion because of one specific and striking feature, such as the del(5) syndrome with polyposis. We comment in greater or 310
•
lesser length upon the genetics of each. In some, we make mention of familial transmission; but primarily we are dealing with de novo defects. The better known ones are depicted in the composite karyotype in Figure 19–7. In the limit, every different deletion, even if only one case is known, could be regarded as a “new syndrome.” As Figure 19–6 attests, from the ECARUCA site, rather a large number of deletions, and an increasing number of duplications, are coming to be recognized. Chromosomal atlases and catalogs provide further clinical information, and the essays in Cassidy and Allanson’s Management of Genetic Syndromes (2005) offer detailed commentaries for some of the more common of these syndromes (Angelman, Prader-Willi, Russell-Silver, Smith-Magenis, velocardiofacial, and Williams syndromes).
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
del 1p36.3 cri du chat
WolfHirschhorn
del 2q37 1
2
3
4
5 WAGR
Williams LangerGiedion 6
7
8 PWS & AS
Rb
13
14
10
RubinsteinTaybi
15
11
12
SmithMagenis
MillerDieker CMT & HPSN
16
17
Alagille
18p-
del 22q11
18q18
9
19
20
21
del 22q13 22
X
Y
FIGURE 19–7 “Composite karyotype” showing the site of the cytogenetic defect in some of the deletion syndromes. AS, Angelman syndrome; CMT, Charcot-Marie-Tooth neuropathy (a duplication); HPSN, hereditary pressure-sensitive neuropathy; PWS, Prader-Willi syndrome, Rb, retinoblastoma plus other features; WAGR, Wilms tumor, aniridia, genital defects, retardation syndrome.
Chromosome 1. del 1q21.1. “New” syndromes, in which the clinical observations are rather nonspecific, or at least variable, are coming to be recognized, and especially as several groups in numerous countries pool their microarray experience. The microdeletion of chromosome 1q21.1 is such an example, and the clinical picture is described in a paper of which the co-authors number 84 (Mefford et al., 2008). Mental retardation, autism, seizures, cardiac defects, and cataract, are included. There may be a hint of a Williams-like facies. Some are de novo cases, some familial with a (usually mildly) affected parent, and some familial from an apparently unaffected parent. 1q21.1 deletion also has the interest of an association with thrombocytopenia-absent radius syndrome (p. 265). A larger deletion embraces a broader phenotype (Velinov and Dolzhanskaya, 2010). del 1p36.3. The del(1)(p36.3) syndrome may be of similar frequency to the del(22q) syndrome, with these two conditions being the most common
syndromes of, respectively, terminal and interstitial deletion. The deletions can be of variable extent (Heilstedt et al., 2003). The clinical features of this syndrome are reviewed in Gajecka et al. (2007) and Battaglia et al. (2008). The facies is variably dysmorphic, and several minor physical anomalies may be observed. The mental defect is usually severe; an unsurprising observation, given that a major brain malformation, perisylvian polymicrogyria, is a characteristic neuroradiological correlate (Dobyns et al., 2008). The segment 1p36.3 is a light-staining region, and thus the deletions, which can be of variable size, are not always visible on light microscopy. In three studies of “karyotypically normal” retarded and dysmorphic populations using subtelomeric FISH, two del(1)(p36.3) cases were identified among a total of 411 individuals tested (Anderlid et al., 2002; Baker et al., 2002; Clarkson et al., 2002). Using an initial molecular approach (a panel of microsatellite markers to pick up 1q36.3 hemizygosity), Giraudeau et al. Structural Rearrangements •
311
(2001) screened 567 patients with mental retardation and found three with del(1p), in whom FISH confirmed the deletion. These combined data suggest that about 0.5% of this category of population, in whom a standard karyotype has been interpreted as normal, may have this subtelomeric deletion, and nowadays most diagnoses are likely to be made through microarray. The majority of cases of del(1) (p36.3) syndrome arise de novo, but transmission from a parental translocation is recorded, as is presumed parental gonadal mosaicism (Shapira et al., 1997b; Gajecka et al., 2008b, 2010). del 1q41q42. This syndrome is another example of discovery by the “genotype-first” approach (Shaffer et al., 2007b). Because certain genes are recognized to cause developmental disabilities, it was presumed that genes within those pathways, not (as yet) shown to have effects on development, might also be candidates for genetic disease. A microarray was constructed that targeted these genes, and among these was the DISP1 locus, a gene that directly regulates sonic-hedgehog (SHH). The SHH signaling pathway, when perturbed, can cause midline brain defects, including holoprosencephaly. Patients were identified with DISP1 loss, and hence the del(1)(q41q42) basis of this loss. Although these patients do not have holoprosencephaly, they exhibit midline defects, have severe mental retardation, and a notable dysmorphology and malformation pattern, including diaphragmatic hernia, reminiscent of Fryns syndrome7 (Kantarci and Donahoe, 2007). Chromosome 2. del 2q23.1. This syndrome, typically de novo, has the interest of a resemblance to Angelman and Rett syndromes, with features of severe mental retardation with absent speech, stereotypic repetitive behavior, microcephaly, ataxia, seizures, and a coarse facies. The crucial genes may be MBD5, a member of the same family as the MECP2 gene of Rett syndrome, and SATB2, these genes both having the capacity to influence expression of multiple other target genes ( Jaillard et al., 2009; Rosenfeld et al., 2009; van Bon et al., 2010; Balasubramanian et al., 2011). del 2q37: Albright-like Syndrome. This cytogenetic defect should specifically be sought in patients with a morphological phenotype somewhat reminiscent of
Albright hereditary osteodystrophy (short stature, short metacarpals), a quite distinctive facies, and intellectual deficit. It may be among the more frequent of the deletion syndromes, the subtlety of the cytogenetic defect having previously obscured its role (Phelan et al., 1995; Wilson et al., 1995). Deletions of lesser extent, detectable only upon molecular methodologies, may display substantially the same clinical phenotype (Kitsiou-Tzeli et al., 2007). Parent-to-child transmission is very rare, but has been observed. Familial translocations have been reported (Batstone et al., 2003). Chromosome 3. del 3p26.2. Differing degrees of distal 3p deletion are associated with differing phenotypes, although a core contributor to the mental retardation is likely to be loss of the gene SRGAP3 (Shuib et al., 2009). Two genes within most 3p deletions are VHL and ITPR1, haplo-insufficiency of which cause Von Hippel-Lindau disease, and spinocerebellar ataxia type 15, respectively (Van de Leemput et al., 2007). One would thus predict that a del(3p) patient whose deletion included these genes, and surviving well into adulthood, might develop each of these tumorassociated and ataxic syndromes. del 3q29. Patients with variable extents of 3q29 deletion, approximately 1.6 Mb in size, have been described by Willatt et al. (2005) and Ballif et al. (2008). Mild-to-moderate mental retardation with microcephaly, and fairly subtle facial dysmorphism, are core features. Some have, in addition, had clefting, and genitourinary malformation. Familial transmission is recorded (Ballif et al., 2008; Li et al., 2009; Digilio et al., 2009). The deletion is flanked by segmental duplications, and likely arises from nonallelic homologous recombination, and thus it is not surprising that the reciprocal duplication is observed (see later, dup 3q29). Chromosome 4. del 4p: Wolf-Hirschhorn Syndrome. This wellknown deletion syndrome identified in the prebanding era is one of the few that can, in its typical form, be confidently recognized clinically. The natural history of Wolf-Hirschhorn syndrome (WHS) is discussed in Battaglia and Carey (1999). Classical deletions are detected on routine cytogenetics,
7 Diaphragmatic hernia has also been associated with deletions at 8p23.1 and 15q26.2, and thus these segments should be specifically interrogated when a diagnosis of Fryns syndrome is being considered clinically (Slavotinek et al., 2005).
312
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
whereas molecular approaches are needed to identify subtler deletions (Maas et al., 2008; Galasso et al., 2010). Zollino et al. (2008) propose that the condition be considered in two typical forms: the more common classical form, and a relatively mild form, and in addition a severe form not resembling classical WHS, and these several forms correlate with the extent of the deletion. Maas et al. discuss the genotype-phenotype correlations, with respect to the precise extent of the deleted segment (as defined by array-CGH), and propose genes whose haplo-insufficiency may be the basis of particular traits. While most WHS occurs de novo, almost half may, upon further analysis with array-CGH, be revealed as having arisen as unbalanced translocations, and they may show additional phenotypic features in consequence (South et al., 2008b; Galasso et al., 2010). The improvement in precision of cytogenetic and latterly the development of microarray methodology has enabled the elucidation of some malformation syndromes previously supposed to have been due to autosomal recessive inheritance. The PittRogers-Danks syndrome turned out to be the less severe phenotypic version of WHS, as noted earlier. A family with the supposed recessive Lambotte syndrome of multiple malformation, having surprisingly produced a further affected child in a succeeding generation, was subjected to detailed FISH analysis, and a familial t(2;4)(q37.1; p16.2) identified (Herens et al., 1997). Here, the phenotype in fact reflected the combined effects of partial 4p monosomy and 2q trisomy. A syndrome of mental retardation with polymicrogyria originally reported as being X-linked transpired as being due to an autosomal translocation, t(1;12)(q44;p13.3), with both adjacent-1 segregations being represented (Zollino et al., 2003). Other examples exist, and Verloes et al. (2000) speak of these as “pseudo-recessive disorders” being revealed in their true colors as due to cryptic translocations. A deletion just centromeric of the WHS region, at 4p15.2-p15.32, leads to a quite different disorder, in some respects resembling Marfan syndrome (Basinko et al., 2008). del 4q34. It may be that among all the chromosomal disorders no single discrete physical sign is pathognomonic for a particular aneuploidy (that is, seen in this specific condition, and in no other). But there is one sign that might be: a duplicated nail of the fifth finger, with one nail in the normal position, and the other where the fingertip pad should be
(thus, dorsal and volar surfaces). This curious observation, it is proposed, can enable the clinician to predict a distal 4q deletion, to be precise, of 4q34.2 (Vogt et al., 2006). A deletion just a little above, into 4q33, may be associated notably (although not pathognomonically) with abnormality of the ulnar ray of the upper limb (Keeling et al., 2001). The range of deletion and phenotype, and consideration also of duplication and more complex distal 4q rearrangement, is documented in Rossi et al. (2009). Chromosome 5. del 5p: Cri-du-Chat Syndrome. The breakpoints in this famous syndrome are very variable: in a molecular study of 62 Italian cases, for example, at least 28 different sites were identified, from p13 through p15.2. The “cri” region is pinpointed to proximal p15.3, and certain other components of the phenotype can be attributed to certain segments within p14, p15.1, p15.2, and p15.3 (Fig. 19–8) (Kjær and Niebuhr, 1999; Cerruti Mainardi et al., 2001). Kjær and Niebuhr suggest that anomalous formation of the notochord in the early embryo may then compromise the development of certain cranial nerve nuclei in the subjacent brainstem, affecting the innervation of the larynx, which is the actual anatomic structure that produces the “cat-like cry.” Deletion of the segment 5p15.3 alone can produce the typical cry in an otherwise normal child. Van Buggenhout et al. (2000) document in quite some detail, with several photographs, the phenotypes in seven older individuals, teenagers and adults. All but one had severe or profound mental defect. Perhaps surprisingly, the neurodevelopmental compromise does not correlate well with the size of the deletion (Marinescu et al., 1999a). Growth charts have been compiled from international data from 374 cases, from birth to age 18 years, and these document a substantial downward shift in the graphs for the three major indices (weight, height, head circumference) (Marinescu et al., 2000). While most cases are sporadic, familial transmission from a parental translocation is recorded (Cotter and Musci, 2001), and this possibility should be checked for in each case. One case of recurrence of del(5)(p15.2), identified at prenatal diagnosis, attests to the reality of gonadal mosaicism (Hajianpour et al., 1991). del 5p13.1: Cornelia de Lange Syndrome. Chromosome 3 had initially been implicated in the cause of Cornelia de Lange syndrome (CdLS), but the true basis was revealed when two affected infants Structural Rearrangements •
313
FIGURE 19–8 An example of the archetypal del(5p) chromosome (above), deletion of which is the basis of cri du chat syndrome, first described in 1963. A display of a detailed karyotype-phenotype correlation is shown below. Regions which may be implicated in speech, the characteristic cry, and facial anomalies are indicated at left. The arrows indicate five different breakpoints defining five different extents of terminal deletion, the lengths of which are reflected in the vertical bars to the right (numbered 1–5), and the associated phenotypes noted alongside. The vertical bar to the left (numbered 6) identifies an interstitial segment, deletion of which does not cause phenotypic abnormality. (From I. Kjær and J. Niebuhr, 1999, Studies of the cranial base in 23 patients with cri-du-chat syndrome suggest a cranial developmental field involved in the condition, American Journal of Medical Genetics 82:6–14. Courtesy I. Kjær and J. Niebuhr; reproduced with the permission of Wiley-Liss.)
were identified with a rearrangement involving 5p13.1. A stillborn infant had a severe form of the condition, amniocentesis in the pregnancy having identified a 5p deletion, 46,XY,del(5)(p13.1p14.2); and another CdLS infant had a translocation t(5;13) (p13.1;q12.1). These cases enabled discovery that the gene causing CdLS is NIPBL—in the great majority due to a point mutation—located at 5p13.1 (Hulinsky et al., 2005; Selicorni et al., 2007). Recurrence due to paternal gonadal mosaicism for a NIPBL mutation is recorded (Niu et al., 2006). Suspected cases testing mutation-negative would prudently proceed to FISH or microarray analysis, to check for whole-gene deletion. del 5q22-q23: Polyposis Plus Syndrome A minor degree of facial dysmorphism and mild to moderate mental retardation are nonspecific features seen in deletions in the region of 5q22-q23; the unique feature is adenomatous polyposis of the bowel, and indeed it was such a deletion that led to discovery of the APC (adenomatous polyposis coli) gene (Hockey et al., 1989; Kobayashi et al., 1991). Absence of one APC allele of itself allows polyps to develop, and any subsequent mutation/loss of the allele on the intact chromosome 5 then leads to loss of the tumor suppressor function of this gene (Hodgson et al., 1994). At the time of initial diagnosis in a child, a cancer risk could not have been anticipated. This exemplifies the point that parents need to be aware that, when a test procedure (microarray) is applied that can interrogate the whole genome, more information may come to light than they had been expecting. Nevertheless, although advice about a cancer risk may come as an “unwanted surprise,” discovery of this deletion (and some others, as noted later) may in fact be lifesaving (Heald et al., 2007). Other similar examples exist of constitutional deletions that convey, in addition to congenital abnormality, a cancer predisposition. As with the polyposis example, the typical scenario is that loss of one allele of a tumor suppressor gene on the deleted chromosome comprises the “first hit” in the process of tumorigenesis. The 13q14 deletion associated with retinoblastoma, noted later, is the classic
example. Jacoby et al. (1997) describe a deletion of 10q22.3-q24.1 in a patient with multiple congenital malformations and juvenile polyposis, the latter presumably reflecting the loss of one copy of the PTEN gene. A 9q22.32q31.1 deletion has been associated with a “Gorlin syndrome plus” phenotype, and an increased risk for cancer is to be expected (see later section on “del 9q22.32q31.1”). We refer on p. 16 to the inadvertent discovery of a p53 deletion, predisposing to Li-Fraumeni syndrome. Carcinogenesis may also be due to gene amplification, and constitutional gain of a particular gene may thus contribute a risk. Willatt et al. (2001b) and Seven et al. (2002) draw attention to the link between constitutional 2p23 duplication and consequent duplication of the oncogene N-myc, and neuroblastoma. We note on p. 16 the risk for leukemia in trisomy 8 mosaicism. del 5q35: Sotos Syndrome. Most Sotos syndrome (cerebral gigantism) is seen with a normal karyotype. A few patients have had a chromosomal abnormality, and since two of these involved translocations with one of the breakpoints at 5q35, and since also some del(5)(q35) cases have had a Sotos-like phenotype, this had suggested there might be a Sotos locus in the region. This hypothesis was subsequently vindicated, the gene in fact being NSD1 (Kurotaki et al., 2002). Most patients have a 2.2 Mb deletion, which encompasses NSD1,8 with nonallelic homologous recombination due to Alu repeats the typical basis of this (Mochizuki et al., 2008). Chromosome 6. del 6q. Three different segments of 6q have been associated with particular sets of clinical traits: del(6)(q11-q16), del(6)(q15-q25), and del(6) (q25-qter). Naturally, with these segments being genetically different, and only on the same chromosome by “evolutionary accident,” the three phenotypic patterns are different, with mental retardation noted as the only universal finding (Hopkin et al., 1997). With respect to the q11-q16 deletions, molecular cytogenetics has since enabled a more precise understanding. Bonaglia et al. (2008) dissect
8 NSD1 has a quite different role in cancer, as one of the two genes at the breakpoints of a somatic translocation t(5;11)(q35;p15.5) that leads to the production of a chimeric protein that may well be an initiating factor in childhood acute myeloid leukemia ( Jaju et al., 2001).
Structural Rearrangements •
315
out the candidate genes in deletions at 6q16.2 associated with a phenotype resembling Prader-Willi syndrome. Moving just 5 Mb centromerically, to 6q16.1, Traylor et al. (2009) report a child whose 2.16 Mb deletion removed only two known genes, EPHA7 (ephrin receptor 7) and TSG1. The reasonable conclusion is drawn, in this latter case, that haplo-insufficiency for EPHA7, a gene whose expression field in the mouse equivalent includes neuronal synapses, has a key role in the functional neurological phenotype of the del(6)(q16.1) syndrome. Another specific deletion, in addition to those noted earlier, is del(6)(q23.2-q24.2), reported in just one case, and concerning a 3-year-old girl whose development was “completely normal to advanced,” and who had been karyotyped as a newborn because of low birth weight (Kumar et al., 1999). While the facies was distinctive, she was said to resemble her family. One might imagine that this particular segment contains no critical brain loci. Chromosome 7. del 7p13: Greig Cephalosyndactyly Syndrome. This acrocephalopolysyndactyly syndrome, classically inherited as an autosomal dominant, is due to mutation at the GLI3 locus. Rare microdeletion cases have haplo-insufficiency of GLI3, as well as loss of some adjacent loci, and the phenotype is combined Greig syndrome with neurodevelopmental defect, seizures, and other abnormalities (Kroisel et al., 2001). del 7p21.1: Saethre-Chotzen Syndrome. Most Saethre-Chotzen syndrome (a type of acrocephalosyndactyly) is due to point mutation in the TWIST gene at 7p21.1. Cytogenetic forms include microdeletions within this region, the larger of which remove other genes and add in a broader phenotype ( Johnson et al., 1998; Touliatou et al., 2007; Busche et al., 2011). An apparently balanced translocation, which might disrupt the TWIST gene or perturb its function, is another mechanism. It appears that either mechanism, haplo-insufficiency or point mutation of TWIST, can lead to the similar phenotype. The skull defect in Saethre-Chotzen syndrome is premature fusion of cranial bones; it is interesting that a duplication (“triplo-excess”) at this locus can produce the opposite effect, an underdevelopment of the cranial bones.
del 7q11.23: Williams9 Syndrome. Williams syndrome (WS) is due to a ~1.5 Mb deletion within chromosome 7q, and can arise from either parent (Wang et al., 1999a). Sperm studies in control donors show similar frequencies of deletion and duplication for this segment, pointing to a likely NAHR mechanism (Molina et al., 2011). The deleted loci include GTF2I and GTF2IRD1, which may well be key contributors to the neurocognitive profile and craniofacial morphology (Antonell et al., 2010; Ferrero et al., 2010), and the elastin gene, which is responsible for the cardiovascular component of the phenotype. The characteristic psychological phenotype is that of a mild intellectual disability, with overfriendliness to strangers, and a lacking in social judgment; an abnormally developed amygdala (a brain structure subserving social behavior and the recognition of emotional facial expressions) may be the basis of these traits (Martens et al., 2009). Earlier impressions that aspects of language might be intact have been refuted (Donnai and Karmiloff-Smith, 2000). Growth indices (height, weight, head circumference) for deletionproven WS have been compiled (Martin et al., 2007b). Affected monozygous twins generally have a rather similar phenotype (Castorina et al., 1997). We know of no record of recurrence in siblings of undoubted WS to normal parents. Rare instances of parent to child transmission are known (Pankau et al., 2001). Atypical cases with larger deletions may show a more severe phenotype, and those with smaller, a less severe. Thus, a child with a large deletion at 7q11.2 had components of WS, along with other features, including a severe mental defect (Wu et al., 1999); and a child with a smaller deletion showed lesser physical traits, and had an IQ within the normal range (Ferrero et al., 2010). del 7q21.3: Ectrodactyly Plus Syndrome. One type of split hand and split foot malformation is associated with deletions in 7q21, and loss of one allele at a “digit-formation locus” in this region may be the basis, and candidate genes have been proposed (Basel et al., 2006). Loss of contiguous genes encompassed by the deletion may contribute to other less specific dysmorphology and to a diminished intellectual function (Roberts et al., 1991). An inversion in this region might have its influence due
9 The custom of removing the apostrophes from the names of authors associated with syndromes has led to the occasional misspelling of this condition as “William syndrome.” Similarly, the terminal s of Edwards and of Sotos is sometimes erroneously dropped.
316
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
to “position effect,” with loss of expression of these putative gene(s), rather than haplo-insufficiency (van Silfhout et al., 2009). Other split hand/foot loci may reside in 2q14.1-q14.2, 4q32-q35, 5q15, and 6q16-q22 (David et al., 2009; Niedrist et al., 2009). del 7q32-qter: Holoprosencephaly Plus Syndrome. Holoprosencephaly is a developmental brain defect that can vary from devastatingly severe to rather mild, and there are several different genetic causes. Chromosomes 7 and 13 are important contributors. Distal 7q deletions were instrumental in the mapping of one locus, HPE3 (Frints et al., 1998), that was narrowed to find the causative gene, SHH. De novo deletion is the rule, but familial holoprosencephaly has been recorded in the setting of a familial 7q36 translocation (Hatziioannou et al., 1991). Deletion 7q holoprosencephaly is due to SHH haplo-insufficiency. Another deletional cause of holoprosencephaly resides in chromosome 13, del(13)(q32), and reflects haplo-insufficiency for the ZIC2 brain morphogenesis gene (Quélin et al., 2009). (This is in contrast to the holoprosencephaly associated with chromosome 13 duplication, as in standard trisomy 13.) In a series of holoprosencephaly patients in whom array-CGH was applied, Bendavid et al. (2009) identified a surprisingly large fraction with de novo microrearrangements, and they proposed that further brain genes might reside in these several regions. Chromosome 8. del 8p23.1-pter, or del 8p23.1. Small terminal deletions of 8p are quite frequently recognized. A notable aspect of the phenotype is a severe behavioral disturbance in childhood, on the background of a mild mental defect. Sudden and extreme changes in behavior are observed, with outbursts of aggressiveness and destructiveness. Frustration tolerance is very low. Behavior seems to improve in later adolescence. The deletion may remove the gene for a cardiac transcription factor, GATA4, and this may be the basis of the observation that heart defects are frequent (Pehlivan et al., 1999). Diaphragmatic hernia may be a characteristic component (Shimokawa et al., 2004). del 8q12: CHARGE Syndrome. Most cases of this syndrome (coloboma, heart, choanal atresia, retarded growth and development, genital, ear
defects) are due to mutation of the CDH7 gene, with functional haplo-insufficiency, but in a few haplo-insufficiency is the consequence of a deletion in 8q12 that includes the CDH7 locus (Vissers et al., 2004). More severe mental retardation in the deletion case may reflect the loss of adjacent “brain genes.” The deletion is typically de novo. del 8q24.11-q24.13: Langer-Giedion Syndrome (Tricho-Rhino-Phalangeal Syndrome Type II). The facies is distinctive, and diagnosis can be made with some confidence on clinical grounds. The condition is due to a deletion that removes the gene for trichorhino-phalangeal (TRP) syndrome type I, a bone growth control gene (EXT1, which causes exostoses10), and several other genes, to give the broader picture of Langer-Giedion syndrome (Lüdecke et al., 1999; Momeni et al., 2000). The deletion may arise on the chromosome 8 of either parent (Nardmann et al., 1997). Chromosome 9. del 9p22-p23. Quite a number of 9p cases involving the p22-p23 region are recorded, over 100, and they present a characteristic phenotype. Many are due to deletions occurring in a region of about 5 Mb in 9p23: not so much a hotspot, but a series of hotspots. The deletion is equally likely to have happened on the paternal or maternal chromosome 9. Some that may at first sight seem to be simple deletions turn out, on FISH studies, to be due to other more complicated rearrangements (Christ et al., 1999; Swinkels et al., 2008). del 9p24.3: Sex Reversal Plus. This deletion syndrome is notable in having pointed the way to discovery of the DMRT1 gene. This is the most conserved of any known sex-determining gene, and it is actually on the Z chromosome (the homogametic chromosome) of birds. Its expression is normally greater in the male than in the female embryo, and this dosage may be the basis of its testis-inducing action. It is proposed that the loss (or perturbation) of one DMRT1 allele in (or adjacent to) the deleted segment brings the amount of product down below this threshold, and thus the 46,XY,del(9)(p24.3) person develops as a female (Calvari et al., 2000). An incomplete loss of function may lead to genital ambiguity. Loss of adjacent genes presumably contributes to the wider phenotype (Vinci et al., 2007).
10 Another deletion syndrome with exostosis, but due to another locus, is Potocki-Shaffer syndrome (see section on “del 11p11.2”).
Structural Rearrangements •
317
del 9q22.32q31.1. This deletion syndrome includes a susceptibility to cancer, due to involvement of the PTCH1 gene, and thus the chromosomal diagnosis enables an informed surveillance to put in place (De Ravel et al., 2009). del 9q34.3: Kleefstra Syndrome. This subtelomeric microdeletion syndrome of severe intellectual deficit and multiple malformation has been observed in over 50 reported cases (Kleefstra et al., 2009) and may be, after 1q36 and 22q13, the third most frequent of the subtelomeric deletion syndromes. In the neonate, the facies may have a resemblance to Down syndrome. Haplo-insufficiency of the gene EHMT1 is, of itself, the basis of the phenotype, and varying degrees of the extent of deletion appear not to influence the clinical picture. Analogous to Rubinstein-Taybi syndrome (see later), patients with classic mendelian mutations in this gene present essentially the same phenotype as do those with deletions. The protein encoded by to this gene, Eu-HMTase1, has a role in maintaining the integrity of histones that comprise a key component of the architecture of the chromosome; this syndrome can thus be considered as a disorder of chromatin remodeling. Parental somatic-gonadal mosaicism with recurrence in offspring has been recognized (Willemsen et al., 2011). Chromosome 10. del 10p13: DiGeorge Syndrome Phenocopy, and “HDR” Syndrome. This deletion is the basis of a condition that may resemble DiGeorge syndrome (DGS; see later section on “del 22q11”), and indeed the condition has been labeled DGS2 (Van Esch et al., 1999), although not every such deletion will lead to a DGS-like clinical picture (Benetti et al., 2009). Some features of “DGS2,” such as ptosis and hearing loss, are not present in the 22q deletion form. It is clearly a rare cause of a DGS phenocopy, as no cases were found in a European study specifically addressing this question, and indeed the authors propose that searching for 10p microdeletions is not warranted in the service laboratory (Bartsch et al., 2003). However, in an American study, in which a dual-probe FISH for DGS1 and 2 was used, one case was found in 412 patients presenting with a possible diagnosis of DGS (54 had DGS1) (Berend et al., 2000b). Hypoparathyroidism is a frequent observation in del(10)(p13), but this is in fact due to haplo-insufficiency for the nearby GATA3 gene, determining the HDR (hypoparathyroidism, sensorineural deafness, renal dysplasia) syndrome, 318
•
when the deletion extends to include this locus (Fujimoto et al., 1999; van Esch et al., 2000; Benetti et al., 2009). Most HDR syndrome occurs sporadically, but inheritance due to parental translocation has been observed. del 10q11.2: Hirschsprung Disease Plus Syndrome. The locus for the receptor kinase gene RET is at 10q11.2 (the Hirschsprung chromosome region 1, HSCR1). In the haplo-insufficient state, certain neurons/neural crest cells may fail to migrate to their proper place and/or fail to undergo proper neuronal maturation in the intestinal wall. Without this nervous control, the segment of bowel is chronically contracted, and this causes a partial or complete obstruction (Hirschsprung disease). The loss of adjacent loci contributes to a wider phenotype (Fewtrell et al., 1994). Deletions in 13q22 and 2q22.3, removing the EDNRB and ZEB2 loci, respectively, are the basis of two other Hirschsprungassociated syndromes (Shanske et al., 2001; Engenheiro et al., 2008); these, and RET, more usually present as classic mendelian mutations. Chromosome 11. del 11p11.2: Potocki-Shaffer Syndrome. In 1993, Shaffer et al. described a family segregating an insertional translocation, with 11p11.2 inserted into proximal 13q, and in which some individuals had the 11p deletion after malsegregation of the translocation. The clinical phenotype included the notable features of multiple exostoses, and craniofacial dysostosis with enlarged parietal foramina, along with mental retardation and micropenis in males. This picture resembled that of a patient with acrocephalosyndactyly and an apparently normal karyotype (Lorenz et al., 1990), in whom the same deletion was subsequently demonstrated. Further such cases have since been recognized, typically due to de novo deletion (Bartsch et al., 1996; Potocki and Shaffer, 1996). The parietal foramina are due to haplo-insufficiency of ALX4, and the multiple exostoses reflect haplo-insufficiency of EXT2 (Wu et al., 2000). An exceptional case is described in Chuang et al. (2005) of a del(11)(p11.2) child whose phenotypically normal mother carried the same deletion, but with a supernumerary 11p11.2 neocentromeric marker chromosome, and thus having an overall balanced genotype. del 11p13: WAGR Syndrome (Wilms Tumor, Aniridia, Genital Defects, Mental Retardation). Haploinsufficiency of the PAX6 morphogenesis gene causes aniridia (absence of the iris). Loss of one
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
WT1 allele can comprise the first hit in the sequence of events to cause Wilms tumor, and it is also responsible for the impairment of genital development. These two genes, along with some putative brain genes, are removed in the 11p13 deletion, and the tout ensemble adds up to the WAGR syndrome (Fischbach et al., 2005; Xu et al., 2008). 11p13 deletions and translocations with a presumed position effect are the cause of a substantial fraction, about 40%, of all cases of aniridia (Crolla and van Heyningen, 2002). Interestingly, a duplication for the segment 11p12-p13 also produces an eye defect, indicating that a PAX6 dosage effect, whether an insufficiency or an excess, influences the morphogenesis of the eye (Aalfs et al., 1997). del 11q23: Jacobsen Syndrome. This syndrome is of some interest because very rarely the fragile site (FRA11B) at 11q23.3 may predispose to the generation of the deletion ( Jones et al., 1994). However, the great majority of patients do not have their deletion breakpoint at or immediately adjacent to the fragile site. Grossfeld et al. (2004) reviewed a large cohort (110 patients), documenting the range of phenotype, and noting the interesting point of a particular type of platelet disorder, associated with neonatal thrombocytopenia, which may be pathognomonic for the syndrome. Van Zutven et al. (2009) review cases due to an interstitial deletion, noting the variability of breakpoints, and including one case of their own due to the mother carrying an insertional translocation 46,XX,ins(4;11)(p14;q24q25). Chromosome 13. del 13q14: Retinoblastoma Plus. The association of retinoblastoma with constitutional 13q– was recognized in the early days of cytogenetics, and this observation gave the clue to the position of the retinoblastoma gene (RB) on this chromosome at 13q14. A wider syndrome can accompany the deletion, including mental retardation and facial dysmorphism, the severity according to the extent of the deletion, those extending distally to q14.1 being of more severe effect than those extending proximally (Baud et al., 1999). One 13q– child is recorded with retinoblastoma detected at age 4 and going on to a diagnosis of Wilson disease, an autosomal recessive disorder of copper transport, at age 11. Since
the Wilson locus is also in this region (13q14.3q21.1), the assumption is that the child’s other chromosome carried a Wilson mutation, and the deletion “exposed” this mutation in the hemizygous state (Riley et al., 2001). While de novo generation is the rule, five cases are known in which a parent carried an insertional translocation involving 13q14 (Punnett et al., 2003). del 13q21.1-qter. A distinct array of malformations is associated with deletions more distally in 13q, and the observations in patients allows karyotype-phenotype mapping of the region (Fig. 19–2) (Kirchhoff et al., 2009). Chromosome 14. del 14q12. 14q12 contains the FOXG1 locus, and loss of this gene as part of a ~4 Mb microdeletion leads to a syndrome of mental retardation and epilepsy, sometimes called “congenital Rett syndrome”; mutation within FOXG1 leads to the similar phenotype. The concomitant microduplication is also recognized (Yeung et al., 2009a; Brunetti-Pierri et al., 2011). Chromosome 15. del 15q11.2. This microdeletion, requiring microarray for its demonstration, has the interest that it encompasses the region between the two alternative proximal breakpoints, BP1 and BP2, of the Prader-Willi/Angelman deletion;11 this particular segment is not subject to imprinting (Bittel et al., 2006). A psychiatric phenotype is frequent. The deletion is often inherited from a less affected parent (von der Lippe et al., 2011). del 15q11-q13.1: Prader-Willi Syndrome, Angelman Syndrome. See Chapter 22. del 15q13.2-q13.3. It is apparent that “brain genes” reside in 15q13.2-q13.3 (very close to, but nevertheless quite distinct from, the Prader-Willi/Angelman region), which may influence intellect, behavior, and the psyche. Autism and poor behavior are particular observations in the individual with a microdeletion, along with variable mental impairment (which can be severe); dysmorphism, if present, is subtle. Other recognized associations are with epilepsy and with schizophrenia. The deletion typically encompasses the segment between two paralogous breakpoint11
11 There are well-defined breakpoints sited on proximal 15q and numbered 1–5 from proximal to distal. Their similarity of sequence, due to low-copy repeats, predisposes to nonallelic homologous recombination between any two of the BPs, with a consequential deletion or duplication of the intervening segment. BPs 1–2 are more proximal, in 15q11.2, and comprise one of the proximal breakpoints responsible for the typical PWS/AS deletion, the distal one being BP3, at 15q13.1. BP 4 is located in 15q13.2, and BP5 in 15q13.3.
Structural Rearrangements •
319
evidence has emerged for a parent of origin effect; furthermore, this segment is not known to be subject to imprinting. Homozygosity leads to a considerably more severe phenotype, with an encephalopathy associated with mental retardation, seizures, and movement disorder, and blindness due to retinopathy (Masurel-Paulet et al., 2010).
(BP) sequences: either the 3.95 Mb segment between BP3-BP5, or, more commonly, the 1.5 Mb segment between BP4-BP5 (and see Table 19–2). A number of cases, including both de novo and familial examples, have been identified in the short period of time since the first recognition, and this may be one of the more common of the microdeletion syndromes (Gu and Lupski, 2008; Sharp et al., 2008; Ben-Shachar et al., 2009; Dibbens et al., 2009; Miller et al., 2009; Pagnamenta et al., 2009; van Bon et al., 2009). But not all individuals are affected, and indeed many family members with the same deletion are normal,12 attesting to an apparent incomplete penetrance of the deletion (Cubells et al., 2011). Thus, this microdeletion may be considered as a predisposing factor, which, in the presence of other as yet undelineated variants, may compromise the integrity of higher functioning of the brain, with varying neurological/psychological/behavioral symptomatology in consequence. A question that gender of the transmitting parent and of the child may be relevant remains open; but thus far, no
In what may be the first example of a genetically targeted pharmacological management in a deletion syndrome, Cubells et al. (2011) report a substantial improvement in aggressive behavior in an adult male with the use of a drug (galantamine) which acts as an agonist for a specific acetylcholine receptor, the gene for which (CHRNA7) is located within the 15q13 deleted segment. It is reasonable to imagine that this drug stimulation enabled maximum activity of the reduced quantum (due to CHRNA7 haplo-insufficiency) of these receptors on brain neurons, and that the neurons thus stimulated allowed an improved
Table 19–2. Deletion and Duplication Syndromes Having Their Basis within Proximal 15q Rearrangement, Listed According to the Breakpoints That Define the Extents of the Deletion/Duplication (and See Note 11) PARENT OF ORIGIN WITHIN 15q11.2-q13 MATERNAL
PATERNAL
Deletion; BPs 1–3, 2–3 Deletion; BPs 1–4, 1–5 Duplication; BPs 1–3
Angelman syndrome Angelman +
Prader-Willi syndrome Prader-Willi +
PDD + autism
Often no effect
WITHIN 15q13.1-q13.3
NO APPARENT PARENT OF ORIGIN EFFECT
Deletion; BPs 3–4, 3–5, 4–5 Duplication; BPs 3–5, 4–5
Variable MR; ASD; neuropsychiatric disorder; minor dysmorphism Variable MR; ASD; neuropsychiatric disorder; minor dysmorphism
ASD, autism spectrum disorder; BPs, pairs of involved breakpoints within this region, numbered 1 through 5; MR, mental retardation; PDD, pervasive developmental disorder. Angelman + and Prader-Willi + refer to more severe forms of these syndromes
12 It may be that some parental abnormality is concealed due to lack of knowledge of the family background. Ben-Shachar et al. (2009) observed rather many of their index cases had been adopted, and they speculate that “the adoption in many cases may have been related to cognitive, psychiatric, and/or social issues in the biological parents.”
320
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
function within the neural substrate that subserves control of behavior. Chromosome 16. del 16p11.2. This chromosomal region is particularly subject to rearrangement, presumably reflecting the existence therein of several duplicons (Bachmann-Gagescu et al., 2010; Barge-Schaapveld et al., 2011). Two major regions subject to deletion in 16p11.2 can be listed: a proximal and a distal segment, the former of ~600 kb, and the latter of ~200 kb. The ~600 kb deletion presents a clinical picture of autism+epilepsy, although the behavioral phenotype may not meet the formal criteria for autism spectrum disorders (Fernandez et al., 2010; Rosenfeld et al., 2010; Shinawi et al., 2010). Macrocephaly is a frequent observation. There may or may not be a mild degree of dysmorphism. The deletion is more usually de novo, but can be inherited. The more distal ~200 kb deletion includes the SH2B1 locus, and this predisposes to severe earlyonset obesity (Bochukova et al., 2010). Larger deletions, of ~7–9 Mb, extend into 16p12.2, and lead to a phenotype which is associated with dysmorphism and mental retardation with severe language disability (but not autism) (Ballif et al., 2007a; Hempel et al., 2009). del 16p12.1. This specific ~520 kb microdeletion, extending from nt 21,854,025 to nt 22,374,785, is another of those that may be nonpenetrant, or less obviously expressed, in a parent, but lead to a phenotype of developmental and language delay and certain physical anomalies, more often craniofacial and skeletal, in the child (Girirajan et al., 2010). The basis of this difference may be that the child has another microdeletion/duplication—a “second hit,” or “second-site genomic alteration”—elsewhere in the genome. Some of these second hits may have sufficed, of themselves, to produce a phenotype, but the picture is more severe in the company of the 16p12.1 deletion. Veltman and Brunner (2010) suggest that this could be a more general mechanism, and that some second hits of this type might simply be additive in their effects, while others, by virtue of involving segments that contain genes in the same developmental pathway as those in the first hit, could lead to an exacerbation of the particular phenotypic traits, rather than adding in new traits. del 16p13.11. This deletion is another to have been discovered “genotype-first,” in five patients from a large cohort screened by microarray analysis (Hannes et al., 2009). It is associated with mental
retardation, microcephaly and epilepsy, and in some patients, short stature, cleft lip, and other midline defects. The typical deletion is 1.65 Mb in size and contains at least 15 genes. del 16p13.3: α-Thalassemia and Mental Retardation. This is one of two α-thalassemia and mental retardation (ATR) syndromes (the other being an X-linked mendelian condition). In the del(16p) ATR syndrome there is monosomy for a >1 Mb segment, including the α chain globin loci and some brain loci (Gibson et al., 2008). A larger deletion determines a broader phenotype, with tuberous sclerosis and polycystic kidney disease as well as the ATR (Eussen et al., 2000). Smaller deletions, in the range 2.7 to 268 kb, rather surprisingly produce no phenotype other than thalassemia, in spite of the deletion of from 1 to 15 other genes (Horsley et al., 2001). Holinski-Feder et al. (2000) report a notable example of ATR-16 due to a subtelomeric translocation, which had escaped detection on FISH, and which only came to light after a pedigree analysis showed linkage to 16p (see Fig. 19–9). del 16p13.3: Rubinstein-Taybi Syndrome. The Rubinstein-Taybi syndrome (RTS) has a distinctive phenotype, and the facies and the broad thumbs are very characteristic. In about one-tenth of cases, the basic defect is a deletion of the gene CBP (cyclic AMP-regulated enhancer binding protein) (Wallerstein et al., 1997; Petrij et al., 2000a). It is, thus, a single locus disorder rather than a contiguous gene syndrome, and point mutation within this gene can also cause the syndrome. The basic defect in CBP leads to a generalized dysregulation of expression in a number of target genes. The deletion can be seen on FISH using cosmid probes (Blough et al., 2000) (Fig. 19–9). There is no obvious clinical distinction between those RTS patients with or without the microdeletion (Taine et al., 1998). The range of observed severity presumably reflects a variable expressivity of the abnormal genotype, and the case of identical twins with RTS having rather different neurobehavioral phenotypes supports this suggestion (Preis and Majewski, 1995). The oldest putative case, from 500–900 CE, is that of a skeleton excavated at the Yokem site in Illinois (Wilbur, 2000); some kind of record would be set were this case ever to yield to a paleocytomolecular genetic analysis! The sites of recombination in the majority of translocation and inversion forms of RTS lie within a breakpoint-cluster region in the 5΄ part of the CBP gene. This region is also involved in somatic rearrangement, and, for example, the translocation Structural Rearrangements •
321
FIGURE 19–9 Two chromosome 16p deletion syndromes, alpha-thalassemia/retardation syndrome (left), and Rubinstein-Taybi syndrome (right). Both deletions are in the distalmost band, 16p13.3, the ATR-16 region being distal to R-T. These deletions are difficult or impossible to see on routine banding, but they are clearly apparent on fluorescence in situ hybridization (FISH) with the appropriate probe, as shown here, with one of the no. 16 chromosomes in each case showing nonhybridization (indicated by the shorter arrows).
t(8;16)(p11; p13.3) can be a contributory event in the genesis of acute myeloid leukemia (Petrij et al., 2000b). The CBP gene thus joins the ranks of the small number of genes known to cause congenital malformation if abnormal during embryogenesis, and cancer if the abnormality is acquired in postnatal life (NSD1 being another; see section on “del 5q35”). del 16q24. Two quite separate syndromes involve band 16q24. First, the del 16q24.1 syndrome is of interest in that it might cast light upon the basis of some cases of the VACTERL (vertebral, anal, cardiac, tracheo-esophageal, renal, limb) association, given that patient observations included these findings, albeit that the phenotype that led to the discovery of the deletion, namely, the severe lung disorder alveolar capillary dysplasia, is not itself a recognized VACTERL component (Stankiewicz et al., 2009). Second, deletion of 16q24.3 leads to a syndrome that includes autism, and thus suggesting that the genes implicated, ANKRD11 and ZNF778, might have a role more broadly in autism spectrum disorders (Willemsen et al., 2010). Chromosome 17. del 17p13.3: Isolated Lissencephaly Sequence and Miller-Dieker Syndrome. A deletion of the brain 322
•
morphogenesis gene LIS1 produces lissencephaly (“smooth brain”), a severe neuronal migration defect, although agyria/pachygyria (absence/thickness of gyri) may be a more accurate description. Deletions are of variable extent and may be intragenic or remove the entire gene (Cardoso et al., 2002). All FISHdetectable deletions confined to LIS1 thus far found have been de novo, implying a very low recurrence risk. 17p lissencephaly is to be distinguished from the other major genetic type, the X-linked syndrome (p. 120). Loss of an adjacent locus or loci, with YWHAE an important gene in this respect, adds in defects of other systems and characteristic dysmorphogenesis, and this constitutes the Miller-Dieker syndrome (MDS) karyotype and phenotype (Nagamani et al., 2009) (Fig. 19–10). The brain malformation is more severe than in the isolated LIS1 deletion, which may reflect the contribution of another brain morphogenesis locus in distal 17p (Cardoso et al., 2003). Some MDS cases have been due to a parental rearrangement (inversion or translocation), not necessarily recognizable on routine cytogenetic testing, and this possibility should be carefully assessed (Yokoyama et al., 1997; Joyce et al., 2002). 17p13.3 deletions that include YWHAE but not LIS1 present different phenotypes (Nagamani et al., 2009).
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
FIGURE 19–10 Miller-Dieker syndrome. Chromosomes showing deletion in distal 17p, at band p13.3. Fluorescence in situ hybridization (FISH) probe does not hybridize to the deleted chromosome (shorter arrow).
del 17p13.1. Deletions in this region may be of differing extents, but a point of common interest is loss of the TP53 gene. This raises a question of susceptibility to Li-Fraumeni syndrome; but, thus far, no such diagnosis has been made (Schluth-Bolard et al., 2010) (and see p. 16). del 17p12: Hereditary Pressure-Sensitive Neuropathy. Hereditary pressure-sensitive neuropathy (HPSN), with alternative names of hereditary neuropathy with liability to pressure palsies (HNPP), and tomaculous neuropathy, is the reciprocal deletion of Charcot-Marie-Tooth neuropathy (see later). The deletion of a particular “nerve gene”—the PMP22, or peripheral myelin protein 22 gene—leads to abnormal myelination of the peripheral nerves, and this compromises their function. A typical presentation is the backpacker who complains of numbness (sensory nerves) and weakness (motor nerves) in the arms after a day’s hiking, and these symptoms are due to the pressure of the shoulder straps on the nerves leading to the arms. Almost all HPSN is due to this type of deletion, and thus detectable using FISH, and by microarray (Fig. 19–11). The deletion can arise de novo, or, as is more usual, can be transmitted from an affected parent. The risk to transmit the defect is 50%. A single family is on record in which the defect was due to a reciprocal translocation, t(16;17)(q12;p11.2), which disrupted the PMP22 gene, and a heterozygous mother and son had HPSN (Nadal et al., 2000).
Two distinct sex-dependent mechanisms are involved that produce the deletion or duplication of the chromosome 17 region associated with, respectively, HPSN and Charcot-Marie-Tooth type IA (Lopes et al., 1998; Inoue et al., 2001a). Rearrangements arising from maternal gametogenesis, which can be either deletions or duplications, are due to an intrachromosomal mechanism, either an unequal sister chromatid exchange or, in the case of deletion, excision of an intrachromatid loop. If the rearrangement occurs in paternal gametogenesis (the more common scenario), it comprises a duplication and arises by unequal meiotic crossing-over between the two no. 17 chromosomes (cf. Fig. 19–1), an interchromosomal mechanism. HPSN is an example of the unusual circumstance of the monosomic state having a less severe phenotype than the trisomic: the neuropathy of CharcotMarie-Tooth disease is more disabling than in HPSN. This might reflect an accumulation within the cell of degraded excess protein in the PMP22 duplication, compromising the cell’s function, versus a mere reduction in the amount of protein with the deletion (Ryan et al., 2002). HPSN is also an example of a deletion that occurs in an otherwise almost gene-bare region. Thus, the phenotype is essentially based upon the dosage effect of just the one gene, PMP22. del 17p11.2: Smith-Magenis Syndrome. The Smith-Magenis syndrome (SMS) encompasses a Structural Rearrangements •
323
FIGURE 19–11 Countertype deletion and duplication in 17p11.2 leading to hereditary pressure-sensitive neuropathy (deletion, left) and Charcot-Marie-Tooth neuropathy (duplication, right). The deletion is evident due to nonhybridization of the appropriate probe on fluorescence in situ hybridization (FISH) of metaphase chromosomes (shorter arrow, left). The duplication can only be discerned when the chromosomes are in their attenuated state in interphase, and two separate (but close) spots identifying the duplicated segment be appreciated (longer arrow, right) in a substantial fraction of cells, while the normal chromosome shows a single spot (shorter arrow).
picture of dysmorphology, mental defect, and fractious behavior (Fig. 19–12). Sleep disturbance (associated in most cases with a reversal of the normal circadian pattern of melatonin secretion, but the neurobiology remains to be clarified; Boudreau et al., 2009) is a characteristic feature; and a habit of
self-mutilation, and a markedly diminished pain sensitivity, can manifest as “onychotillomania” (pulling out nails) (Smith et al., 1998a; Potocki et al., 2000b). To the practiced eye, the facies may be distinctive (Allanson et al., 1999). Most (>90%) patients have a ~4 Mb deletion, resulting from an
FIGURE 19–12 Smith-Magenis syndrome. Chromosomes showing deletion in proximal 17p, at band p11.2. Fluorescence in situ hybridization (FISH) probe does not hybridize to the deleted chromosome (shorter arrow). This region is proximal to the segment deleted in hereditary pressure-sensitive palsy (Fig. 19–11). 324
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
unequal meiotic crossover followed by nonallelic homologous recombination, the stage having been set by the existence of flanking low-copy repeats (Fig. 19–1); a larger deletion may be associated with a more complicated phenotype (Natacci et al., 2000). The crucial locus within the deleted segment is apparently the retinoic acid induced protein 1 (RAI1), haplo-insufficiency of which may compromise the activity of a number of downstream genes, and each of these, thus compromised, then contributing to a component of the syndrome (Girirajan et al., 2009). No instance is known of recurrence in a family of SMS due to the common deletion. del 17q21.31. This is a condition that could, in retrospect, be seen as a syndrome, but in which the collection of features did not impress sufficiently that recognition was likely to have been achieved ahead of the laboratory discovery (“genotype-first”) of this recurrent deletion. Poor speech development, epilepsy, and a friendly disposition have led some to see a resemblance with Angelman syndrome. There are only four definite genes in the deleted region; and it remains to be learned whether this is a contiguous gene syndrome or essentially a single-locus disorder, the remaining genes being dosage insensitive (Grisart et al., 2009; Tan et al., 2009). del 17q23.1q23.2. This is another genotype-first syndrome, in which nonallelic homologous recombination is the usual basis of the microdeletion (Ballif et al., 2010). Heart and limb defects are characteristic, which may be due to haplo-insufficiency of the transcription factor genes TBX2 and TBX4. Chromosome 18. del 18p, del 18q. Quite substantial deletions of chromosome 18 short arm and long arm were recognized in the very early days of medical cytogenetics; the small size of this chromosome facilitated recognition of these deletions on “solid-stain” cytogenetics (De Grouchy et al., 1964; De Grouchy et al., 1966). Two-thirds of 18p deletion cases are due to a de novo simple terminal deletion, the remainder reflecting de novo rearrangement, or malsegregation of a (possibly cryptic) parental rearrangement (Wester et al., 2006; Turleau 2008). Familial cases are on record (Maranda et al., 2006). The phenotype has been refined, and attempts made at a clinical correlation according to the nature of the deletion; sophisticated studies may be necessary to distinguish simple terminal deletions from more
complex rearrangements. A very small (subtelomeric) 18p deletion may produce a less severe physical and functional neurological phenotype, but including important psychopathology (BabovicVuksanovic et al., 2004). One particular locus in the 18q deletion whose haplo-insufficiency may contribute to the mental defect is MBP (myelin basic protein) at 18q23. Gay et al. (1997) studied twenty 18q– patients with brain scanning, in order to determine how well the white matter of the brain was developed. In 19 patients in whom the MBP gene was included in the deletion, reduced myelination was demonstrated, whereas in the one patient whose deletion did not include MBP, a normal appearance of the cerebral white matter indicated that myelination had proceeded, at least grossly, without compromise. Array-CGH has enabled a more precise genotypephenotype correlation of this dysmyelination, and certain other associated traits (kidney malformation, aural atresia, failure of response to growth hormone, and has also increased our understanding of the variability of the underlying rearrangements (Cody et al., 2009; Heard et al., 2009). Chromosome 19. del 19p13.3-pter; del19p13.13; del19p13.12; del 19q13.11. Chromosome 19 is gene-dense (Color plate Fig. 3–10), and it is not surprising that this chromosome has been little represented in the ranks of deletion syndromes. Nor is it surprising that these microdeletions are small, and require subtelomeric FISH or array-CGH for their recognition and delineation, as Archer et al. (2005), Malan et al. (2009), Schuurs-Hoeijmakers et al. (2009), Dolan et al. (2010), and Bonaglia et al. (2010) illustrate, with respect to the four recorded syndromes, del(19) (p13.13), del(19)(p13.12), del(19)(p13.3-pter), and del(19)(q13.11). Yet further variations upon this theme come from very small deletions, such as the interstitial 174 kilobase deletion in 19p13.3 described in de Smith et al. (2011), which, while very likely causative, it cannot yet definitely be assumed that the associated phenotype is due to the detected imbalance. Chromosome 20. del 20p12: Alagille Syndrome. The characteristic features of this syndrome are stenosis of the peripheral pulmonary arteries, and insufficient development of bile ducts within the liver (thus, “arteriohepatic dysplasia”), along with certain eye Structural Rearrangements •
325
and skeletal defects, and a distinctive facies (Krantz et al., 1997). Most patients have a mutation in the gene JAG1, but a very few have a cytogenetic defect. In one series of 109 subjects, only three had visible chromosomal abnormalities: two deletions, and one unbalanced 4;20 translocation (Crosnier et al., 1999). The phenotype did not vary between those with whole-gene deletions and those with point mutations, suggesting that haplo-insufficiency is the common mechanism. The first report of preimplantation diagnosis by microarray of an Alagille deletion due to a parental t(2;20)(q21;p12.2) appears in Treff et al. (2011b). A single case report exists of transmission from a mosaic parent (Laufer-Cahana et al., 2002). A more extensive deletion can encompass the region for Wolff-Parkinson-White syndrome, a disorder of cardiac conduction, the key locus being BMP2. The deletion of both JAG1 and BMP2 produces a syndrome combining the defects of each (Lalani et al., 2009). del 20q13.33. This deletion is yet another that was formerly missed on classical cytogenetics, and subsequently revealed on microarray (BériDeixheimer et al., 2007; Kroepfl et al., 2008). In the case of Kroepfl et al. of a dysmorphic and mentally retarded child, the deletion was further delineated on FISH analysis using 20q-derived BAC clones, and then with quantitative RT-PCR interrogating the region of the two breakpoints. Thus, and as we are coming to see more frequently, the description achieves the ultimate degree of precision: a deletion extending from one defined nucleotide position to another, in this case, from nt 62,209,440 to nt 62,301,971. This is a segment of only 92,531 nucleotides, and thus of the two genes contained therein (MYT1 and PCMTD2), one or both can be taken as the causative basis, due to being in the haploinsufficient state, of the clinical phenotype. BériDeixheimer et al. (2007) describe two cases of de novo 20q13.33 deletion. Detailed mapping was performed by microarray CGH in one patient, and confirmed by FISH in both patients. They propose, based on their cases and review of the literature, two clinically distinct phenotypes: one with mild mental retardation, and the other a more complex and severe phenotype. Chromosome 21. del 21q: Partial Monosomy 21. There are 21q deletions of varying degree, and a genotypephenotype exercise allows an assessment of the 326
•
contribution of different haplo-insufficient segments to the observed range of phenotypes (Roberson et al., 2011). Deletion of the segment encompassing the APP (amyloid precursor protein) and SOD1 (superoxide dismutase-1) loci is particularly important (Chettouh et al., 1995). Most cases are sporadic, but some have occurred in the setting of a parental balanced translocation (Huret et al., 1995). (Full monosomy 21 has, in the past, been reported; but restudy with more powerful methodology has shown these to be, in fact, partial 21 monosomies due to unbalanced translocations; West and Allen, 1998.) Chromosome 22. del 22q11.21-q11.23: 22q11 Deletion Syndrome. Before their common cytogenetic basis (Fig. 19–13) was understood, the 22q11 deletion syndromes had a number of labels, including DiGeorge syndrome (DGS), velocardiofacial (VCF) syndrome, and Shprintzen syndrome (De Decker and Lawrenson, 2001). DGS was the name typically applied to a child with heart defect, parathyroid abnormality, and immunodeficiency; in Shprintzen syndrome a cleft or deficient palate was the notable feature; while VCF syndrome emphasized the facial appearance, along with palatal (“velo”) clefting and a heart defect. What used to be called Kousseff syndrome we can now see as 22q11 deletion with the phenotype including neural tube defect (Forrester et al., 2002), and the Cayler syndrome of asymmetric crying facies plus cardiac outflow defect is another variation on the theme (Akcakus et al., 2003). In true acknowledgment of the first definition of the syndrome, in the Czechoslovakian literature in 1955, Sedlácková syndrome may be the most fitting (Turnpenny and Pigott, 2001). With a birth incidence of about 1 in 4000, this is the most common human site of deletion, and this vulnerability likely reflects the nature of low-copy repeats in the 22q11 region. The minimal deletion size is ~1.5 Mb, with some being up to ~3 Mb in extent. Zori et al. (1998) studied a group of patients presenting with velopharyngeal insufficiency, of whom 30% proved to have a deletion, and 13 medical geneticists who studied full-face photographs at a meeting achieved only a 62% correct assignment to del(22q11) or not; repeating the exercise in the 2010s might yield a better result, as the subtleties of the clinical picture become better appreciated. Mehraein et al. (1997) suggest that about a quarter of children with an “isolated” complex cardiac malformation who test positive for a 22q11 deletion in
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
FIGURE 19–13 Two chromosome 22q deletion syndromes, 22q11 (left) and 22q13 (right). The DiGeorge critical region probe identifies the proximal 22q11.21 deletion (left). One chromosome 22 homolog shows normal hybridization (longer arrow), while the deleted chromosome fails to hybridize (shorter arrow). Arrowheads show control probe. The distal 22q13.33 deletion (right) is indicated by the smaller arrow.
fact show other features of the phenotype (especially the facial/endocrine/immune components), if these are carefully sought. Isolated clefting is not associated with the deletion (and neither with the duplication) (Sivertsen et al., 2007). Psychiatric disorder can be associated, and bipolar disorder and schizophrenia are particular concomitants (Swillen et al., 2000); this may relate to the loss on the deleted chromosome of the microRNA processing gene, DGCR8 (Stark et al., 2008). Some motor neurological dysfunction (drooling, abnormal speech, dysphagia) may result from a specific failure of development of the motor cortex of the brain (Bingham et al., 1997). In the familial case, a parent can, for example, show mild features of the condition, or have a predominantly Shprintzen facial and palatal phenotype, with a child showing a characteristic DGS cardiac and endocrine phenotype (Devriendt et al., 1997). There seems not to be a clear correlation between the extent of the deletion and the clinical phenotype, and the fact that most monozygous twins are discordant is a perplexing observation (Amati et al., 1999; Singh et al., 2002b). It may be that there is not one universal DGS gene, but that various combinations of haplo-insufficiencies can lead to a similar, or a not so similar, clinical picture. Attractive candidates as haplo-insufficient genes include TBX1, a transcription-factor gene, YPEL1, a craniofacial morphogenesis gene, and GNB1L, of unknown function (Farlie et al., 2001; Paylor et al., 2006; Prasad et al., 2008; Scambler, 2010).
Most cases are de novo, but about 10% are said to be inherited. It may be that earlier estimates of a larger fraction of affected parents were biased due to studying more remarkable families (Swillen et al., 1998). Indeed, Smith and Robson (1999) report only 5% of parents to have the deletion in an Australian series of 59 cases. Typically, 22q– parents show poor social functioning, and some have frank psychiatric disease. Presumed parental gonadal mosaicism has been described, and prenatal diagnosis using FISH in a subsequent pregnancy will cover this possibility (Sandrin-Garcia et al., 2002). Since the condition is not rare, occasional instances will happen of more than one case in a family, purely coincidentally (Saitta et al., 2004). De novo generation can presumably be invoked in the very rare instance of complementary del/dup mosaicism, as Dempsey et al. (2007) describe in a patient with an atypically small 22q11.2 segment of 1.5 Mb (see also earlier section on “Complementary Deletion/ Duplication”). del 22q11.2. A rare syndrome due to deletion just beyond the typical 22q11.21-q11.23 site presents a somewhat different clinical phenotype, in one case resembling Goldenhar syndrome; this deletion is due to nonallelic homologous recombination of different LCRs than those involved in the classic syndrome mentioned earlier (Ben-Shachar et al., 2008; LafayCousin et al., 2009). All cases have been de novo. del 22q13.3: Phelan-McDermid Syndrome The particular trait is a failure to develop expressive Structural Rearrangements •
327
language, and high pain tolerance is also notable; the physical phenotype comprises fairly “soft” dysmorphism. Phelan (2008) provides a detailed description. Most cases may be sporadic, with deletions of size varying from 100 kb to 9 Mb. Inherited translocations and inversions have been identified, and an instance of parental mosaicism. As well as simple deletions, rings (p. 207) and translocations can be the basis (Luciani et al., 2003). Of the contiguous genes involved, loss or compromise of SHANK3 (also called proSAP2) is likely to be an important contributor to the phenotype (Bonaglia et al., 2006). The diagnosis has been made incidentally when a distal 22q internal control probe (ARSA), used at the time of diagnostic testing for the 22q11 deletion, failed to hybridize. Bonaglia et al. discuss the technical difficulty in detecting the deletion, especially when very small, and the advisability of having a low clinical threshold for suspicion, in any child with failure of speech acquisition. An intriguing suggestion is that intranasal insulin may be useful in improving behavior and cognitive capacity (Schmidt et al., 2009). Chromosome X. X deletions detectable on classical cytogenetics are discussed in Chapter 13. del Xp22.3: X-Linked Ichthyosis. Deletion of the steroid sulfatase (STS) gene at Xp22.3 leads to a pre- and a postnatal phenotype, expressed in placenta and skin, respectively. With a male fetus, the pregnancy is likely to proceed beyond full-term, and there may be the need to induce labor; and in early childhood, the boy develops extensive ichthyosis. A low or absent maternal serum unconjugated estriol (uE3) level in pregnancy is a strong indication of STS deficiency. In one study (Kashork et al., 2002), of nine mothers with low/absent uE3, prenatal diagnosis in six identified (in the male fetus) a complete STS deletion on FISH, and in one, a partial deletion; these seven mothers were all shown to be heterozygous. The reciprocal duplication of the STS locus is a benign population variant in both males and females. del Xp22.3: X-Linked Kallmann Syndrome. Failure of normal pubertal development, and an inability to smell, are the hallmarks of Kallman syndrome (KS). The KAL1 gene at Xp22.3 controls the migration of two classes of neuron to their proper place in the hypothalamus: neurons that would normally produce a hormone (GnRH) to trigger the pituitary to release sex hormones, and neurons in the olfactory 328
•
pathway. Only a minority of KS is X-linked and due to the KAL1 locus (other loci are autosomal), and of these, most are point mutations. So only infrequently will a FISH-detectable Xp deletion be found. Deletions of wider extent may include the steroid sulfatase (see earlier, del Xp22.3) and ocular albinism loci, producing a wider clinical picture (Hou et al., 1999). Small duplications of KAL1 are found in the population as a normal variant. DU PL I CAT I O N S
While there are certainly very many individual duplication cases on record, rather fewer duplication phenotypes have acquired eponymic (or acronymic) status than with deletions, and we do provide a somewhat shorter catalog than the listing of deletion syndromes earlier. Some have a countertype deletion, listed earlier (and see Table 19–1); in the case of those with no recorded reciprocal deletion, it may be that haplo-insufficiency is nonviable. Chromosome 1. dup 1q21.1. The phenotype of this microduplication includes, as does the deletion for the same segment (see earlier), mental retardation and autism (Mefford et al., 2008). As the microdeletion leads to microcephaly, so does the microduplication cause macrocephaly (Brunetti-Pierri et al., 2008). Chromosome 2. dup 2p22p25. A locus or loci controlling the process of neural tube formation apparently resides in 2p24. Duplication that includes this region may be associated with neural tube defect (NTD), spina bifida or anencephaly, and perhaps the duplication map in Figure 3–11 (in Chapter 3) should be updated in this respect. 2p24 may be the most focused “NTD region” in the karyotype, although a few other putative chromosomal sites of NTD susceptibility are recorded (Thangavelu et al., 2004). dup 2q37. The 2q37 duplicated state may be associated with intellect within the normal range, and little or no dysmorphism (Batstone et al., 2003), in contradistinction to the deletion (see earlier). Chromosome 3. dup 3q29. The reciprocal duplication of the 3q29 microdeletion syndrome (see earlier) has the notable feature that the majority of cases are familial, in contrast to the deletions, which arise de novo (Ballif et al., 2008; Lisi et al., 2008). As with the deletion,
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
mild to moderate mental retardation and microcephaly are the most commonly observed traits in index cases. The normality of a transmitting parent, which is sometimes observed, may reflect an incompletely penetrant genotype, and other genetic factors may influence expression, as per the concept discussed in Veltmann and Brunner (2010) earlier with respect to the del 16p12.1. Chromosome 7. dup 7q11.23. This duplication is the reciprocal recombination product to the Williams syndrome (WS) deletion (see earlier section on “del 7q11.23”). Familial cases have been observed. The phenotype is variable, with expressive language delay (in contrast to the loquacity of WS) as a primary component (Orellana et al., 2008; Torniero et al., 2008); this trait was more severely manifest in a child with a triplication of the WS segment (Beunders et al., 2010). Chromosome 10. dup 10q24: Split Hand Foot Malformation Syndrome Type 3. Tandem duplication of the 10q24 region, preferably using array-CGH for its recognition, has been associated in a minority (about 18%) of cases presenting with this malformation syndrome (Everman et al., 2006). One example of sibship recurrence due to maternal somatic-gonadal mosaicism is on record (Filho et al., 2011). Chromosome 15. dup 15q11q13.1: Syndrome of Intellectual Impairment and Autism without Dysmorphism. Duplications of the proximal region of 15q, between breakpoints 1 and 3 (Table 19–2), may determine a syndrome of intellectual impairment, ranging from borderline to severe (“pervasive developmental disorder”), with autism often a prominent feature (Battaglia, 2008; Hogart et al., 2009). Physical findings are usually unremarkable. The frequency may be about 1 in 600 in a referral population with “developmental delay” (Thomas et al., 2003b). The typical duplication includes, but is not as extensive as, the chromatin present in the large idic(15) (see earlier), and the phenotype apparently reflects this, in being not quite as severe as with the idic(15) (Torrisi et al., 2001). The Prader-Willi/
Angelman critical region (PWSCR/ASCR) is contained within the duplicated segment, a point of practical usefulness in laboratory analysis. FISH analysis typically shows two spots on the abnormal chromosome, although in some instances molecular analysis is necessary to prove the point (Thomas et al., 2003a). These dup(15)s have the same breakpoints as in the deletions of the Prader-Willi and Angelman syndromes (PWS, AS), involving misalignment of the same duplicons, and thus representing the countertype of the PWS/AS deletions; both inter- and intrachromosomal mechanisms are implicated, and the process may occur on either parental homolog ( Ji et al., 2000; Roberts et al., 2002; Hogart et al., 2009; Molina et al., 2011). (Note that this dup(15) is not to be confused with the 15q11.2-q13 euchromatic variant due to constitutional cytogenetic amplification, which does not involve the PWSCR/ASCR, and which is nonpathogenic; p. 264). A duplication between breakpoints 1 and 6 leads to a more severe phenotype (KitsiouTzeli et al., 2010). As with the 15q11q13 deletion syndromes (that is, PWS and AS), there is a parent-of-origin effect: the syndrome characteristically results when the duplication is transmitted from a heterozygous mother (or if de novo, it is of maternal generation), but not when it is paternally transmitted (Yardin et al., 2002; Thomas et al., 2003a). A biological correlate of this observation is that the UBE3A gene is expressed at a greater level from a maternal-originating duplication (Herzing et al., 2002)13 A very few cases of abnormal children with a duplication of paternal origin may reflect there having been an atypical imprinting in the region; or the observation may be coincidental. Given that the phenotype is essentially confined to brain functioning, this is the tissue of particular study relevance. Hogart et al. (2009) measured quantitative reverse transcription-polymerase chain reaction (RT-PCR) transcripts from postmortem material in two dup(15)(q11q13) individuals. In tissue from a male with severe cognitive impairment and seizures, they showed that gene expression and DNA methylation correlated with parental gene dosage. However, in a female with autism and milder PWS-like traits, unexpected deficiencies were seen in paternally expressed transcripts of SNRPN, NDN,
13 Intriguingly, the subset of autistic children manifesting the trait of “insistence on sameness” show greater maternal sharing of alleles in the 15q11q13 region (Shao et al., 2003).
Structural Rearrangements •
329
HBII85, and HBII52, and the levels of maternally expressed UBE3A were unchanged. Their findings suggest that genetic copy number changes, combined with additional (epi)genetic or environmental factors, influence clinical heterogeneity in the 15q11q13 duplication syndromes. Parental studies are appropriate, in order to enable accurate advice about recurrence, in particular distinguishing de novo from familial, and in the latter, maternal versus paternal origin. dup 15q13.2q13.3: Syndrome of Neuropsychiatric Disorder and Autism. As noted in the “Deletion” section, this rearrangement involves a segment very close to, but distinct from, the 15q11-q13.1 segment discussed earlier (Table 19–2). The clinical picture with the duplication is similar to that of the deletion, predominantly one of intellectual impairment, difficult behavior, poor attention, autistic features, and subtle dysmorphism. Counseling is complicated by the incomplete penetrance and variable expressivity of this genomic disorder (Miller et al., 2009; van Bon et al., 2009). dup 15q26qter: Syndrome of Overgrowth and Intellectual Disability. A growth control locus, the insulin-like growth factor receptor type 1 (IGFR1), is sited within distal 15q, at band q26. Duplication of this locus is associated with a syndrome of which overgrowth is a particular feature, along with macrocephaly and a degree of intellectual compromise (Faivre et al., 2002; Nagai et al., 2002). This contrasts with the growth retardation that characterizes deletions of this region (e.g., the ring 15 syndrome, see p. 205). From an analysis of four (inherited) cases, Tatton-Brown et al. (2009) delineated the clinical features in some detail, noting a distinctive facial appearance, and that renal anomalies may be frequent. Chromosome 16. dup 16p11.2. This microduplication is characterized by attention-deficit disorder and poor behavior, speech delay, and microcephaly; dysmorphism, if present, is mild. Parental transmission is frequent, and some relatives may be apparently unaffected (Rosenfeld et al., 2010). dup 16p13.3. This countertype of the RubinsteinTaybi syndrome (RTS) deletion leads to a recognizable phenotype, and like RTS, the key gene whose
duplication is substantially responsible for the clinical picture may be CBP (Marangi et al., 2008; Dallapiccola et al., 2009). In a series of 12 cases in Thienpont et al. (2010), 10 were de novo, and 2 came from an unaffected parent, pointing to an occasional nonpenetrance. dup 16p13.11. Albeit that Hannes et al. (2009) concluded that no phenotype resulted from the 1.65 Mb duplication that is the reciprocal of the deletion (see earlier), Williams et al. (2010) observed an association between a somewhat smaller (824 kb) duplication and attention-deficit hyperactivity disorder, and they refer also to an association with schizophrenia. Reports such as these are likely to spur further CNV studies in these and other psychiatric and behavioral disorders, and the counselor will need to keep a weather eye open. Chromosome 17. dup 17p13.3. Duplication of the “Miller-Dieker region” leads to a less severe phenotype than does the deletion (Roos et al., 2009). Although there is (completely unsurprisingly) mental retardation, there is no gross neuronal migration malformation. One child had a normal brain scan at age 4 years; another infant had a possibly increased signal on scanning in the periventricular region (which is the site of origin of the cortical neurons), and a hypoplastic corpus callosum. dup 17p12: Charcot-Marie-Tooth Neuropathy. The most common form of Charcot-Marie-Tooth neuropathy (CMT1A) is due to the duplication of about 1.7 Mb in 17p12, which encompasses the PMP22 (peripheral myelin protein 22) gene (Nelis et al., 1999).14 It is the countertype of the deletion (see earlier), which causes pressure-sensitive neuropathy. The duplication leads to the production of a 150% amount of the PMP22 protein, and this excess mars the capacity for proper functioning of the peripheral nerve. The major effect is on the motor nerves, and weakness is the important consequence. The nerves to the peroneal muscles (on the outside of the leg, with tendons passing around the ankle to the foot) are particularly vulnerable, and an alternative name for the condition is peroneal muscular atrophy. The laboratory diagnostic test was initially based upon FISH, which had the benefit of giving a direct
14 The duplicated segment of chromosome 17 is some 1.7 Mb in size, but it is “gene-sparse.” The very few other genes contained therein appear to imply no phenotypic consequence due to their being in imbalanced state (as also noted earlier in the deleted state; see section on “del 17p12”).
330
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
visual demonstration of the duplication, with probe hybridizing twice to the duplicated chromosome, and seen as two adjacent fluorescent spots, with the third spot due to fluorescence from the other chromosome appearing elsewhere in the nucleus (Shaffer et al., 1997a). Now, MLPA is used in a number of laboratories; and we expect that, in due course, targeted microarray will become the routine approach. (Other genetic forms of CMT are of course not recognized by these tests.) An intriguing example is that of a de novo X-autosome translocation 46,X,der(X)t(X;17) (p22.1;p11.2) in a mildly retarded female who had CMT (King et al., 1998). The extra segment of 17p attached to Xp produced an attenuated picture of partial 17p trisomy, presumably reflecting an extension of inactivation into the 17p segment from the X-inactivation center of the der(X). The PMP22 gene on the 17p segment was apparently fully functioning, however, since the neuropathy was typical for CMT. The inactivation process could be supposed to have “jumped over” the PMP22 region (this process discussed on p. 117). Another rare circumstance is that of homozygosity for the duplication: effectively, a partial 17p tetrasomy. This leads to a more severe manifestation of the neurological phenotype (Pareyson et al., 2003). Lebo (1998) proposes that prenatal diagnosis should be made available for CMT, notwithstanding that CMT can be a relatively mild handicap, and comments: “Given the slow rate of progress toward curing all forms of human genetic disease, patients with degenerative diseases who already have irreversible nerve pathology should not be offered undue hope for intervention by gene therapy.” Couples will make their own decisions. dup 17p11.2: Potocki-Lupski Syndrome. This condition has the interesting history that it was predicted to exist, as the reciprocal recombination product of the Smith-Magenis deletion (see earlier) (Potocki et al., 2000a). The prediction was vindicated. The clinical picture is less severe than in Smith-Magenis syndrome, and it includes such rather nonspecific features as mental retardation, infantile hypotonia, and failure to thrive (Potocki et al., 2007). dup 17q21.31. The reciprocal of the deletion for this segment (see earlier) leads to a syndrome of microcephaly with severe cognitive compromise and difficult behavior with autistic features, along with fairly mild facial dysmorphism (Kirchhoff et al., 2007; Grisart et al., 2009). Thus, the duplicated state
is not notably less severe than in the deletion, as is otherwise quite often observed. This rearrangement (deletion and duplication both) is associated with a particular inversion polymorphism in the parent that facilitates nonallelic homologous recombination between low-copy repeats flanking the region. Thus far, all cases have been de novo. Chromosome 22. dup 22q11.2. The countertype of the common del(22q11) is a duplication for the same segment (Ensenauer et al., 2003; Portnoï, 2009). Theoretically, the dup(22q11) should be similarly common, but it is not—or to be precise, it is not commonly recognized. The clinical picture is quite diverse and may include, in common with the deletion, velopharyngeal insufficiency; a specific association may be bladder exstrophy (Draaken et al., 2010; Lundin et al., 2010). It may comprise no more than abnormal behavior and indeed may merge into normality; and thus it is advisable that parents of diagnosed children be karyotyped (Yobb et al., 2005). Brunet et al. (2006) looked at a large group of patients displaying clinical features of the 22q11 deletion syndrome, to determine whether any 22q11 duplications might be found. None were; although, naturally enough, they did discover some deletion cases. dup 22q13. The countertype of PhelanMcDermid syndrome presents with developmental delay, very limited language acquisition, and mild dysmorphism (Okamoto et al., 2007). Chromosome X. dup Xp11.22p11.23. This duplication, seen in a number of sporadic and familial cases, and for the most part affecting the sexes similarly, presents a picture of mental retardation with a largely nondysmorphic physical appearance, but it does display a characteristic pattern on the electroencephalogram (Froyen et al., 2008; Giorda et al., 2009) (and see p. 229). dup Xq22: Pelizaeus-Merzbacher Disease. This is a disease of the white matter of the brain and presents a severe neurodegenerative clinical picture. The genetic defect resides in the PLP (proteolipid protein) locus at Xq22. Proteolipid protein is a major structural component of myelin in the central nervous system. A quite common mutational basis of Pelizaeus-Merzbacher disease is duplication (or occasionally triplication) of about a megabase of DNA at Xq22 (Woodward et al., 1998). This is analogous to Charcot-Marie-Tooth neuropathy type 1A Structural Rearrangements •
331
(see earlier): an additional copy of a gene that produces myelin, but in this case the myelin that sheathes of axons of the central nervous system. While maternal mosaicism is known, most mutations arose from the (unaffected) maternal grandfathers of affected boys, and it thus appears that an intrachromosomal event taking place in male gametogenesis is the usual source of the defect; the mechanism of “fork stalling and template switching” (p. 296) may underlie the generation of the rearrangement (Lee et al., 2007). As with CMT 1A, interphase FISH in Pelizaeus-Merzbacher disease may show adjacent spots on the X, reflecting the tandem nature of the duplication, although molecular methodology may be taking over as the mainstay of diagnosis, including prenatal diagnosis and carrier assessment (Warshawsky et al., 2006). dup Xq27.1: X-linked Panhypopituitarism. An important gene in the development of the pituitary gland is SOX3, the locus of which resides in Xq27.1. Some families with X-linked panhypopituitarism (the pan referring to involvement of all the component parts of the gland) are due to duplications of Xq27.1, with the additional dose of SOX3 the presumed cause of the abnormal gland development. In some families, the duplication is large enough to be seen on high-resolution cytogenetics, and these larger duplications are associated with the additional observation of mental retardation (Woods et al., 2005). dup Xq28: Lubs Syndrome. A microduplication in distal Xq28, in the region including the MECP2 and FLNA genes, leads to a syndrome of severe mental retardation with a recognizable facies (a narrow, “pinched” nose being typical), and which may include the functional neurological complications of mixed hypotonia/spasticity, and bowel and bladder dysfunction (Clayton-Smith et al., 2009). The duplication can be transmitted by unaffected females, in whom skewed X-inactivation may be protective. Recurrent breakpoints are defined (Bartsch et al., 2010).
recurrence risk applies. The possibility of occult parental gonadal mosaicism warrants consideration of prenatal diagnosis. SU P ER N UM ER A R Y M A R K ER C HR O M OS O M ES : 4 7 , + ma r
Parental mosaicism is unlikely, but not completely excluded, if the parental lymphocyte karyotypes are normal. Although a risk for recurrence will be “small” (which we cannot precisely define, although we presume a low single-digit percentage figure, possibly <1%), prenatal diagnosis may be appropriate. I SO CH R O M OS O M ES (NONACROCENTRIC)
A couple having had a child with an isochromosome, for a chromosome other than an acrocentric, can generally be given encouraging advice, more especially if the child is mosaic. The major mechanisms of generation are considered to operate either at meiosis II or postzygotically, and in either case no discernibly increased risk would be implied. Exceptions to this (noted in the earlier “Biology” section), presumably due to parental gonadal mosaicism, are extremely rare. The unbalanced “Robertsonian isochromosome” is a different category and is to be seen in a different light (Chapter 7). Triplication. With very few cases reported, an empiric recurrence risk cannot usefully be determined for the typical sporadic case. Parental gonadal mosaicism remains a possibility (perhaps even a probability), as illustrated by the example in Eckel et al. (2006) of a phenotypically normal mother with a 46,XX,trp(12)(pter→p11.22::p11.22→p12.3: :p12.3→qter)[6]/46,XX[44] karyotype, her son having the triplication in nonmosaic form. Such an example obliges caution. At the microarray level, rare instances are reported of a nonpathogenic duplication in a parent leading to a pathogenic triplication in his or her child (p. 267).
I NT E R C H RO MOS O M A L RE A RRA N GE M E N T WITH DUPLICATION: 46,rea
The great majority of these rearrangements arise de novo, presumably from illegitimate meiotic recombination between nonhomologs, and a low (<½%)
332
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
20 chromosomal disorders of sex development
THE IDEA of a female with XY chromosomes and a male with XX chromosomes may seem a contradiction in terms. Yet to those who have studied the mechanisms of sex development, perhaps what is more remarkable is that most of the time there is a clear association between being XX and female, and being XY and male. The XX and XY embryo are built on a fundamentally similar outline plan, and only as development proceeds do certain modifications evolve. If at any point in this sequential process some genetic instruction is faulty, inappropriate, or cannot be acted on, the direction of sexual development may proceed imperfectly. In the extreme, the opposite path is taken. This latter state is the particular subject of this chapter, with more of a focus upon those forms in which classical and molecular cytogenetics comprise the key diagnostic investigations, although we do touch on some mendelian conditions. We provide categories for “girls and women,” and “boys and men,” according to the phenotypes presented, and the sex that the individual is regarded
as being, by the individual or by the individual’s parents. N OMEN CL AT UR E
These conditions are subsumed under the general heading of disorders of sex development (DSD) (Hughes, 2008). The different chromosomal categories may be indicated by reference to the sex chromosome constitution (XX or XY) and the nature of the gonad (testis, ovary, ovotestis, or dysgenetic/ streak). The former expressions XX male, XY female, and hermaphrodite are now referred to as particular types of DSD. Genital ambiguity is now simply denoted XX DSD or XY DSD, according to karyotype; clearly these are rather broad descriptors, and more precise detail might usefully be added in individual cases. With reference to male or female sex, these different levels of definition can apply: gonadal sex (ovary, testis, ovotestis, streak); anatomical/ genital sex (structure of the internal and external
•
333
genital tract); karyotypic sex (46,XX, 46,XY, or other); and behavioral sex (gender identity).
BIOLOGY Somewhat simplified, the fundamental plan of the reproductive tract is that bilateral gonads, arising from the genital ridge, connect with bilateral paired internal ducts (müllerian and wolffian), which enter a midline genital sinus, opening at the perineum. This opening is buttressed on each side by labioscrotal folds and capped above by a phallus. The basic plan of the genital ridge is laid down according to instruction from, in particular, the WT1 and SF1 genes. Thereafter, the direction in which gonadal development proceeds is due to the activity of a number of genes on the sexual differentiation pathway.
The Key Role of the SRY Gene In the absence of SRY, but with input from WNT4 and RSPO1, the gonad develops into an ovary, and the duct system develops into fallopian tubes and uterus. The genital sinus remains as an opening (the vagina), flanked and surmounted by labia and clitoris. The female state results. If a Y chromosome is present—or at least that part of the Y that contains SRY, the testis-determining gene—the male direction is taken. The SRY gene calls into action, among others, the SOX9, FGF9, and MAP3K1 genes, and the gonad becomes a testis. The testis, in turn, secretes hormones, of which androgen influences the genital tract to masculinize, and anti-müllerian hormone causes regression of the female müllerian ducts. A vas deferens forms from the duct system. The phallus enlarges. The labioscrotal folds fuse in the midline and accommodate the descending testes. The male state results.
DISORDERS OF SEX DEVELOPMENT IN GIRLS AND WOMEN XY Ovarian Disorder of Sexual Development The 46,XY karyotype in an otherwise normal girl, with (apparently) completely normal female anatomy, is a very rare observation. A single case is reported of a child who was a compound heterozygote for mutations in the CBX2 gene, discovered 334
•
only because of a discordant chromosome finding at prenatal diagnosis (Biason-Lauber et al., 2009). The internal genital tract was normal female, the gonads of normal ovarian appearance, and normal upon histology. It may be that SRY requires activation by CBX2 before it can make its male-determining contribution to sexual differentiation.
XY Disorder of Sexual Development, Complete Pure Gonadal Dysgenesis (Swyer Syndrome) The rare familial form provides a unique example of a mendelian condition that can be inherited in an X-linked recessive, Y-linked, or sex-limited autosomal dominant or recessive mode. In the X-linked forms or autosomal forms, the XY female has a perfectly normal Y chromosome, with a normal SRY testis-determining gene; presumably, there is a mutation in a gene (whether this be X-linked or autosomal) controlling a later event in the testicular developmental pathway. One such autosomal gene has been discovered: the DHH gene, of which the locus is at 12q12. Of six apparently nonconsanguineous Mexican-mestizo patients with this form of XX DSD, three were homozygous for a DHH mutation, and two of whom, not known to be related, had the same mutation (Canto et al., 2004). (The same gene has been implicated, in heterozygous state, in mixed gonadal dysgenesis; Canto et al., 2005.) In the Y-linked form, there is a mutation in the SRY gene itself. In some Y-hemizygotes, the mutant gene has nevertheless been able to reach a threshold of operation and to induce testis development, while in others with the same mutation it has not. Thus, for example, an XY male with a mutation in SRY may be a normal fertile man, while his XY child may be a daughter. The threshold is apparently all or nothing: partial expression, that is to say an intersex state, does not result ( Jäger et al., 1992; Imai et al., 1999). A man may be a gonadal mosaic for an SRY deletion, as presumably was the father in Barbosa et al. (1995). Two sisters with this form of XY DSD (one with gonadoblastoma) had a deletion of SRY, but their father showed a normal SRY result; there were three other normal sisters and six normal brothers. Similarly, Schmitt-Ney et al. (1995) describe two XY sisters and their XY half-sister with an SRY point mutation, whose father was shown to be mosaic for this mutation.
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
Sporadic occurrence is usual, and in about 15% of these cases the SRY gene has a de novo mutation, or rearrangement, that abolishes its function of testis determination. XY females with an intact SRY gene might plausibly have an abnormality of one of the other genes, autosomal or gonosomal, in the testisdetermining pathway. Candidates include the genes mentioned earlier (WT1, SF1, WNT4, RSPO1, SOX9, FGF9, MAP3K1, CBX2, and DHH) and also DAX1, DMRT1, SOX3 (the “ancestor” of SOX9), and TSPYL1 (Hughes, 2008; Vinci et al., 2009). In the fullness of time, we may anticipate that a “DSD chip” will be developed, to test for these several genes; Barbaro et al. (2008) have produced an MLPA kit to interrogate the dosage of a number of relevant genes. The gonad in this form of XY DSD is dysgenetic, and it is seen as a “streak.” The genital tract feminizes. The lack of female sex hormones causes failure of normal pubertal development. Amenorrhea and failure of pubertal development are the usual complaints that lead these girls to seek medical advice. Gonadoblastoma, a premalignant neoplastic change in the dysgenetic gonad, is common, and it may progress to dysgerminoma (Verp and Simpson, 1987; Berg et al., 1989; Lukusa et al., 1991; Uehara et al., 1999a). Familial ovarian malignancy was a notable observation in a sibship of three XY women (the karyotype presumed in two who had died at ages 19 and 20) described in Kempe et al. (2002). As always, an accurate diagnosis is needed to give useful counseling. It is thus disconcerting that, in a review of 48 women carrying a diagnosis of “XY female,” undertaken in a specialist clinic, in only half was the description accurate. In about onethird of women, the diagnosis was inaccurate, and in one-eighth, frankly wrong (Minto et al., 2005). Counselors will want to assure themselves that the information they have about a patient is correct, and they should painstakingly review all the test findings, with appropriate expert advice.
XY Disorder of Sexual Development, Complete Androgen Insensitivity Syndrome This is a mendelian condition, in which the locus happens to be on the X chromosome. Here, the defect lies further down the developmental path. The gonad becomes a testis and produces testicular secretions, but the genital tract, internal and external, is resistant to the effects of androgen. The inheritance
is X-linked recessive, and the locus is the androgen receptor gene at Xq11–12 (Brinkmann, 2001). The individual appears externally very much as a female, but there is amenorrhea and pubic and axillary hair is absent. Internally, the vagina is short, and the uterus and tubes are represented only by remnants; the testes may be in the inguinal canal (Boehmer et al., 2001). Malignancy of the gonad, gonadoblastoma or dysgerminoma, is less of a concern, occurring in a minority, in 9% in one study, although in two series no cancers were found. About 5% may be an average figure; the risk may be more so if the gonad is undescended (Lukusa et al., 1991; Rutgers and Scully, 1991; Collins et al., 1993; Alvarez-Nava et al., 1997; Chen et al., 1999b). One example is on record in which, in a sense, the X-linkage was directly visible to the cytogeneticist: that is to say, the X chromosome was abnormal, including the region containing the androgen receptor locus. An affected aunt and niece had the karyotype 46,Y,inv(X)(q11.2q27) and the connecting mother 46,X,inv(X)(q11.2q27) (Xu et al., 2003). A unique case is that of androgen insensitivity due to uniparental disomy X in a woman with the XXY karyotype (Uehara et al., 1999b).
DISORDERS OF SEX DEVELOPMENT IN BOYS AND MEN XX Testicular Disorder of Sexual Development Most males with 46,XX testicular DSD (“XX males,” in former parlance) arise from the presence of Yp material (rarely visible cytogenetically) on one of the X chromosomes (Rigola et al., 2002), from occult XX/XXY mosaicism, or from the inappropriate activity of a gene that is normally switched on only in response to a Y-originating genetic instruction. In about three-quarters of cases the SRY gene is present, typically the consequence of an abnormal exchange between the X and Y during meiosis I in gametogenesis in the father, and thus clearly a sporadic event (Weil et al., 1994; Wang et al., 1995). These are referred to as SRY + XX males, or SRY + XX testicular DSD. The phenotype in SRY+ XX testicular DSD is similar to that of Klinefelter syndrome, presumably reflecting the similar basic genotypes of active X+ inactive X+ SRY in the two conditions; however, the male with XX testicular DSD differs in being of Chromosomal Disorders of Sex Development •
335
normal height and of unimpaired intelligence (Ferguson-Smith et al., 1990). Margarit et al. (1998) describe six SRY + cases due to translocation of Yp material to Xp22.3, in whom different Y breakpoints could be identified, but whose clinical phenotypes were very similar: normal intelligence, normal stature, and testicular atrophy with azoöspermia. In these SRY + males with XX testicular DSD, a more accurate cytogenetic designation would be 46,X, der(X)t(X;Y)—or more fully 46,X,der(X)t(X;Y) (p22.3;p11.2), albeit the exchange is not usually visible on standard cytogenetics—and so there is reference to this entity also in the section on the X;Y translocation (Chapter 6). Rare cases are known of a male with XX testicular DSD in whom the SRY gene had been translocated onto a terminal arm of an autosome (Dauwerse et al., 2006; Queralt et al., 2008). Males with XX testicular DSD and having no SRY gene are denoted SRY–. The fact of male development being able to proceed (to some extent, at least), despite the lack of SRY product, presumably reflects an inappropriate activation of the testisdetermining cascade in an otherwise normal 46,XX embryo, either as a sporadic stochastic event or due to some genetic predisposition. Concerning the latter, Jarrah et al. (2000) report an extended inbred kindred in which XX individuals with varying degrees of masculinization were present, and they suggest that in this family SRY – XX maleness and XX ovotesticular DSD represented a continuum of the same disorder (and see later section on “Ovotesticular DSD”). Grigorescu-Sido et al. (2005) describe an SRY – XX testicular DSD case with imperfect masculinization, whom they contrasted with the normal maleness of two SRY+ XX men. Rare cases may reflect an abnormal dosage of another gene in the sex-determining pathway; SOX9, RSPO1, and SOX3 have been implicated in this respect (e.g., Parma et al., 2006). Three cases are reported of males with 47,XXX chromosomes. In one well-studied example, the man was mildly intellectually disabled, with gynecomastia and hypogenitalism, and severe testicular atrophy on biopsy (Ogata et al., 2001). One X of the three was positive for SRY. In addition to an Xp-Yp interchange in paternal gametogenesis that produced the SRY-positive X chromosome, a coincidental maternal nondisjunction was responsible for
a disomic X ovum. Thus, the combination at fertilization was XX(mat) + der(X)t(X;Y)(pat), giving 47,XX,der(X)t(X;Y) and appearing karyotypically as “47,XXX.” XX testicular DSD has been diagnosed prenatally, following the recognition that the chromosomal and ultrasonographic anatomical genders did not match (Trujillo-Tiebas et al., 2006). (This should however be a carefully considered diagnosis, at least for sporadic cases, as mostly the XX “chromosomal sex” will be due to culture of maternal cells.) 45,X M A L E
We refer to this rare disorder on p. 134. Most, quite possibly all, “45,X males” have, in fact, a molecular translocation of the SRY gene to an autosome or to the X chromosome (and might therefore be thought of as a type of Y;autosome or X;Y translocation). In some, the underlying constitution might actually be an X/XY mosaicism. Y I S O CH R O M OS O M ES
A Y isochromosome, idic(Y)(q11), in mosaic state with a 45,X line, is a rare observation in individuals presenting with a disorder of sex development1 (Lungeanu et al., 2008). These chromosomes presumably arise in paternal gametogenesis, with loss in an early mitosis or mitoses of the embryo, to produce the 45,X line.
Ovotesticular Disorder of Sexual Development The term hermaphroditism, of classical Greek derivation, is losing some favor among those so diagnosed; and the qualifiers “true” and “pseudo” were always somewhat arcane. These days, we speak of ovotesticular DSD. As that descriptor indicates, the defining feature is that the gonads comprise both ovarian and testicular elements: there may be a testis and an ovary, or one or both may be an ovotestis. The most common karyotype is 46,XX (thus, XX ovotesticular DSD), seen in 60%; one-third have mosaicism with one cell line which includes Y chromosomal sequences, mostly 46,XX/46,XY; a few are 46,XY; and other more rare forms are known (Krob et al., 1994; Queipo et al., 2002). Ovotesticular DSD
1 But the usual presentation with the idic(Yq) is infertility in an otherwise normal male (p. 391).
336
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
generally presents as a problem in determining the sex of a newborn infant (Hadjiathanasiou et al., 1994). Most of the 46,XX cases test negative on peripheral blood analysis for the SRY gene, and in some of these the basis of the defect may be sporadic inappropriate activation of the testicular developmental cascade in part of the gonadal tissue during its embryonic formation. Alternatively, an apparent XX karyotype may harbor Y material, as Margarit et al. (2000) show in a woman reared as a boy with hypospadias who went on to have gender change surgery after testing “46,XX.” Several years later, reanalysis revealed a tiny segment of Yp translocated on to the X long arm, 46,X,der(X),t(X;Y)(q28;p11.31). A more common explanation in the 46,XX case may be cryptic mosaicism within the gonad itself, with an island or islands of tissue containing the SRY gene (Ortenberg et al., 2002; Queipo et al., 2002). It is a curious and unexplained fact that ovotesticular DSD (mostly with a 46,XX karyotype) is far more common in the South African black population than in Europeans (Wiersma, 2004). The XX/XY state more usually results due to the fusion of twin XX and XY embryos (XX/XY chimerism). Strain et al. (1998) reported a notable example of iatrogenic ovotesticular DSD, which followed in vitro fertilization, presumably due to an XX and an XY embryo fusing; Malan et al. (2007) reached a similar conclusion in a case diagnosed prenatally, and which could be referred to as “tetragametic chimerism.” Another mechanism is that an ovum might divide symmetrically (instead of budding off a polar body), and the two cells are each fertilized by a sperm (Chen et al., 2005c). A further theoretical route is from the postzygotic loss of the X and of the Y in separate cells of an initially 47,XXY conception (Niu et al., 2002). The basis may be molecular, rather than cytogenetic. For example, one nonmosaic 46,XY case had a postzygotic mutation in SRY with SRY+/SRY–gonadal mosaicism (Braun et al., 1993). Presumably the SRY+ line was responsible for the testicular elements in the gonad, and the SRY – line for the ovarian elements. A somewhat similar patient is described in Modan-Moses et al. (2003), in whom 46,XX SRY+/45,X SRY+/45,X SRY–mosaicism was associated with a clinical picture of ovotesticular DSD. Other mosaicisms include XX/XXY and X/XY. Kanaka-Gantenbein et al. (2007) report a boy, regarded as normal except for an undescended left testis, who presented as a 13-year-old with a left scrotal hemorrhage. In fact, the undescended gonad
was an ovary, which had actually ovulated, and presumably this had been the cause of the bleed. There was a left hemi-uterus and fallopian tube; the testis, on the right, was dysgenetic. On both blood and testicular biopsy the karyotype was 46,XX/47,XXY; as expected, given the presence of a male gonad, albeit an imperfect one, SRY and AZF loci were present. In another case, a baby girl presenting with clitoral hypertrophy typed 46,XY on blood, but analysis of the removed dysgenetic gonads revealed X/XY mosaicism (Röpkea et al., 2007). On histological examination, the gonads contained testicular and ovarian elements: the XY state was observed more in the testicular component of the gonad, while cells with only an X predominated in the ovarian fraction. A single case is recorded of ovotesticular DSD associated with an autosomal cytogenetic abnormality, and this may reflect the effects of an autosomal gene on the cascade of sexual differentiation (Aleck et al., 1999). This child had ambiguous genitalia, with one ovarian and one testicular gonad, and karyotyped 46,XX,rec(22)dup(22q)inv(22) (p13q13.1)mat. Testing for SRY was negative. Tomaselli et al. (2008) report the first actual autosomal mutation to be recognized, in the RSPO1 gene, in a woman with ovotesticular DSD, and concomitant palmoplantar hyperkeratosis. This gene is located at 1p34.3; the mutation was in homozygous state, and her parents were first cousins. Rare familial 46,XX cases of ovotesticular DSD may reflect a mutation, whether autosomal or X-linked, such as RSPO1 or others in this pathway, that induces the testis developmental cascade to proceed at a post-SRY stage. Slaney et al. (1998) describe the case of four 46,XX cousins with abnormal sexual differentiation. Three had 46,XX ovotesticular DSD, and one was a 46,XX male. The putative testis-development gene had been transmitted through two mothers. Affected distant relatives due to a familial X;Y translocation are noted on p. 135. Sterility is almost universal. But of the 11 pregnancies to women with ovotesticular DSD reviewed in Schultz et al. (2009), extraordinarily, all infants were male. The opposite applied to the 46,XY man with ovotesticular DSD reported in Zayed et al. (2008), who had had surgery for removal of an intraabdominal testicular seminoma, and which included ovarian elements. At the same operation, a uterus and tubes were identified, and removed. A few years later, he underwent testicular aspiration of the remaining gonad, which yielded sperm: these were Chromosomal Disorders of Sex Development •
337
used for ICSI, and eventually a pregnancy resulted— and a normal daughter was born. RARE D IS O RD ER S WI T H E X TRAG O N A DAL D E F EC TS
A number of rare conditions exist in which sex reversal coexists with physical, metabolic, and in some, mental defect. By way of example, one of these is XY female DSD campomelic dysplasia (campomelia refers to long bone bowing) with sex reversal. The usual cause is a mutation within the SOX9 gene (at 17q24.3q25.1), one of the genes operating on the sexual differentiation pathway and which also influences limb bud mesenchymal development (Wagner et al., 1994). A cytogenetic form of this syndrome is seen in an apparently balanced 17q translocation, which may lead to deletion with haplo-insufficiency, or inactivation, of the SOX9 locus (see Fig. 19–5 in Chapter 19). In the other direction, a chromosomal imbalance may lead to male genital development in the setting of an XX gonosomal complement. Thus, Seeherunvong et al. (2004) describe a child with a chromosome 22 duplication, 46,XX,dup(22)(q11.2q13), who was SRY-negative and had male external genitalia, with intrascrotal gonads, the only genital abnormality being a first-degree hypospadias. The child also had a number of dysmorphic features and psychomotor retardation. A mendelian condition is XX male DSD with palmoplantar hyperkeratosis, in which homozygosity for RSPO1 mutation is the basis (the same gene noted earlier with reference to one form of ovotesticular DSD) (Parma et al., 2006). Microarray methodology can reveal the agency of a sex-determining gene in a DSD individual. A 46,XY girl with a syndrome of ovotesticular DSD, absent uterus, and clitoromegaly, along with skeletal anomalies and developmental delay, had a microdeletion of 3 Mb at 9q33.3, and one of the genes within this segment is SF1. Point mutations in this gene (and as mentioned earlier) are otherwise on record in patients with XY DSD. In this girl, haplo-insufficiency of the protein product, steroidogenic factor-1, is the likely primary basis of the gonadal defect. Haplo-insufficiency for another gene in the deleted segment, LMX1B, led to the disordered skeletogenesis, and a clinical diagnosis of genitopatellar syndrome was made (Schlaubitz et al., 2007).
338
•
GENETIC COUNSELING XY Disorder of Sexual Development, Complete Gonadal Dysgenesis (Swyer Syndrome) Familial/Inherited Cases. XY DSD with gonadal dysgenesis, when familial, is mostly inherited as an X-linked recessive, or autosomal dominant or recessive with expression limited to the XY state. Autosomal recessive inheritance would be improbable in a multigenerational family tree, while on the other hand, this mode would be strongly supported in a single affected sibship with more than one affected, and in the setting of parental consanguinity. In the multigenerational scenario, a clear interpretation of autosomal versus X-linkage may not be possible. The risk to the female carrier (as judged by position in the pedigree) to have an affected child would be a simple 25% if the X chromosome is implicated, but not readily calculable if a partially penetrant autosomal gene is the cause. Although the XY female phenotype is close to that of a normal female, but of course associated with infertility, some couples may want to consider prenatal diagnosis. The use of cytogenetics (XY) and ultrasound morphology (female external genitalia) would presumably allow detection of the condition. As noted earlier, DSD genes are coming to be identified (e.g., DHH in some XY DSD), and genetic counseling will be better underpinned as this knowledge evolves. The Y-linked form is recognized by the demonstration of an SRY mutation carried by the XY girl and her XY father. This circumstance would allow the counselor the rare opportunity to apply principles of Y-linked inheritance with incomplete penetrance to risk estimation. Mutational analysis of the SRY gene (including deletion detection) may provide the basis for carrier detection and prenatal or preimplantation diagnosis. Sporadic Cases. Advice on the recurrence risk in the sporadic case is less straightforward. If a de novo SRY mutation is demonstrated, only paternal testicular mosaicism—which, as noted earlier, has been observed—could imply an increased risk for recurrence. The DHH gene as the basis of an autosomal recessive form is mentioned earlier. Again, prenatal diagnosis by chromosomal/ultrasound gender discordance should be feasible.
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
The rare syndromes of XY female with extragonadal defects need to be judged on their individual merits. For the XY woman herself, assisted conception is possible if a uterus is present, and a handful of successful pregnancy outcomes, using donated gametes, have been reported (Kan et al., 1997; Dirnfeld et al., 2000; Selvaraj et al., 2002; Ko et al., 2007). Aspects of Management. Couples electing not to consider prenatal diagnosis (or to continue a pregnancy in which a positive diagnosis has been made) should know of the importance of two factors in managing these girls. First, the psychosexual orientation of these individuals is female. But with secondary sexual characteristics developing incompletely, and infertility being invariable, their selfimage is vulnerable. In discussing the condition with parents, the counselor should note the importance of using language that reinforces their view of themselves as girls and women, and the counselor should avoid using such terms as “genetic male.” It may be explained to them, beginning in simple terms in childhood, that a genetic factor prevented their ovaries from developing normally (Goodall, 1991). Pregnancy may be achievable with in vitro fertilization using a donor ovum, as noted earlier. Second, there is a substantial risk of neoplastic change in the dysgenetic gonad. A gonadoblastoma arises in around half of familial XY gonadal dysgenesis. The gonadoblastoma itself is noninvasive, but it is often associated with malignant elements, most commonly dysgerminoma, which do invade. Thus, and given that the gonad does not usefully contribute in terms of hormone production, early (first decade) gonadectomy is advisable (Troche and Hernandez, 1986; Verp and Simpson, 1987; Lukusa et al., 1991). Considerable publicity in 2009 concerning an athlete who recorded extraordinary times in women’s running races at an international meeting put in sharp focus the question of how such people are to be regarded. The unfortunate woman’s internal genital state (which apparently included testicular elements) became the subject of public speculation and then of public documentation. The Athletics Federation attempted a resolution, acknowledging her “unfair” but entirely innocent physical advantage as a runner, in allowing her to keep her gold medal, but also awarding a gold medal to the secondplaced athlete.
XY Disorder of Sexual Development, Complete Androgen Insensitivity Syndrome This condition is inherited as an X-linked recessive trait, and the risk of recurrence follows classic mendelian principles. The carrier may be identified and preimplantation/prenatal diagnosis accomplished by molecular analysis of the androgen receptor gene. While complete androgen insensitivity typically has a consistent phenotype within families, allowing for good prediction of the consequences of recurrence, incomplete androgen insensitivity can have variable phenotypes within a family (Boehmer et al., 2001). Issues relating to prenatal diagnosis are discussed in Morel et al. (1994), who also make the interesting but unsurprising point that incomplete forms can imply a worse burden than the complete form, with partially virilized males (known as Reifenstein syndrome) having “considerable psychological distress and poor function in their adult life.” Similar considerations with respect to gender orientation in the XY girl, as discussed in the preceding section, apply to complete androgen insensitivity. The risk for neoplastic change in the gonad is less, in the vicinity of 5%, in the case of complete androgen insensitivity syndrome (CAIS). Thus, some propose gonadectomy may reasonably be delayed to allow spontaneous pubertal feminization ( Jones, 1978; Verp and Simpson, 1987), although regular clinical and imaging checks would be advisable.
XX Testicular Disorder of Sexual Development Many XX testicular DSD boys are not diagnosed until after childhood, by which time the parents are likely to have completed their family. Some cases may be recognized at amniocentesis following discordant karyotypic and ultrasonographic sex. The great majority occur as sporadic events in a family, and in these the likelihood of recurrence is very small. If the child is SRY +, and the father’s X is SRY –, sporadic occurrence is proven. As for the rare case of the SRY – XX male, once the postulated gene or genes have been identified, those cases that would carry a high recurrence risk will be able to be identified. If prenatal diagnosis is requested, and the fetus is 46,XX, testing for SRY along with an ultrasound assessment of external genital morphology should enable distinction (Ginsberg et al., 1999).
Chromosomal Disorders of Sex Development •
339
Ovotesticular Disorder of Sexual Development The considerable majority of ovotesticular DSD represent sporadic cases, these being characterized by a 46,XX karyotype and absence (at least on peripheral blood analysis) of the SRY gene, and presumed to reflect an “accidental” activation of the testis-determining cascade during gonadogenesis, or cryptic intragonadal mosaicism, as discussed earlier. In some cases, the cytogenetics (46,XY, 46,XX/46,XY, or other mosaic karyotype) or molecular genetics (SRY mutation that is not present in father) may allow a more secure reassurance of nonrecurrence. Recurrence is very rare; but a
340
•
positive family history would, of course, imply a high risk, and the RSPO1 gene could be a candidate (see earlier discussion; a dermatological phenotype may aid diagnosis). In the SRY – form, a handful of families are described in which there is also a sib with XX male syndrome, and these cases may speculatively reflect “leaky mutations” in a gene operating at a point downstream in the cascade of sexual differentiation. A familial X;Y translocation is dealt with on its merits. If the condition is diagnosed prenatally, and the pregnancy continued, counselors should consult Hughes et al. (2006) for a consensus statement on management of children with DSDs.
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
21 chromosome instability syndromes
A DEFECT OF DNA repair is the factor underlying the chromosome instability syndromes, also known as chromosome breakage syndromes (Brewer et al., 1997; Michelson and Weinert, 2000; Taylor, 2001). The “instability” refers to the predisposition of the chromosomes to undergo rearrangement or to display other abnormal cytogenetic behavior. Their inclusion in this book is warranted in that special cytogenetic techniques may have a role in clinical diagnosis and prenatal diagnosis, albeit that direct molecular analysis is having an increasing role, as more is learned of the mutational basis of these syndromes. The classic chromosome instability syndromes are Fanconi pancytopenia syndrome, Bloom syndrome, and ataxia-telangiectasia. The main cytogenetic features are listed in Table 21–1. They are mendelian conditions, and in each the mode of inheritance is autosomal recessive. There is genetic heterogeneity in Fanconi syndrome, with cells homozygous for one mutation able to correct in vitro cells homozygous for another mutation
(“complementation”). We briefly note three other rare mutagen-hypersensitivity syndromes—the Nijmegen breakage syndrome; the immunodeficiency, centromeric instability, facial anomalies (ICF) syndrome; and Seckel syndrome. Proneness to cancer is a common concomitant of several of the breakage syndromes (Duker, 2002). Some of these genes have in common their interaction with the breast cancer susceptibility gene BRCA1, their protein products forming a “BRCA1-associated genome surveillance complex” (Futaki and Liu, 2001). Rare or even unique families with various clinical presentations have been associated with chromosomal instability, and some representatives are mentioned in this chapter. Chromosome instability has been reported as an occasional observation in quite a number of known conditions. This list includes, among others, the Cockayne/cerebro-oculo-facialskeletal syndrome spectrum, xeroderma pigmentosum, Rothmund-Thomson syndrome, Dubowitz syndrome, and Riyadh chromosome breakage syndrome. But in several the associations are not clear, •
341
Table 21–1. The Three Classic Chromosome Instability Syndromes CYTOGENETIC FEATURES
Fanconi Increased spontaneous and pancytopenia inducible chromosome breakage AtaxiaIncrease in chromosome breaks, telangiectasia presence of clones with translocations having specific breakpoints in 7, 14, and X Bloom Increased spontaneous and syndrome inducible SCE; increased spontaneous chromatid breakage with production of symmetrical quadriradials the relevance for genetic counseling is uncertain (other than in supporting a diagnosis), and we do not consider them here. Likewise, chromosome instability is a feature of many cancers, and it may indeed be a crucial factor in the process of carcinogenesis; but this a somatically acquired attribute, and not of relevance in the present context. A different cytogenetic observation is that of premature sister chromatid separation. This is a feature of Roberts syndrome, Cornelia de Lange syndrome, variegated aneuploidy syndrome, and Warsaw breakage syndrome, and we make brief mention of these conditions. The genes underlying these disorders code for cohesins, which contribute to the control of sister chromatid segregation at cell division, and thus are dubbed “cohesinopathies.”
CLINICAL GENETICS AND CYTOGENETICS The three classic chromosomal breakage syndromes, as well as Roberts syndrome, Nijmegen breakage syndrome, and the ICF syndrome, are of autosomal recessive inheritance, and the recurrence risk, for parents who have had one affected child, is 1 in 4. In those rare instances in which parenthood is achievable, the risk to the child will in most cases be very low. Cornelia de Lange syndrome is almost always due to a de novo mutation.
Fanconi Pancytopenia Syndrome This uncommon disorder of protean manifestation (also known simply as Fanconi anemia [FA]) is the least rare of the breakage syndromes (Tischkowitz and Hodgson, 2003; Kennedy and D’Andrea, 2005). Originally described as a disorder of short stature, characteristic facies, and certain malformations along with progressive bone marrow failure, the picture has now widened. In one-third of FA there are no major congenital malformations, although many of these will have minor anomalies, skin pigmentary abnormalities, microöphthalmia, and growth indices below the 5th centile (Giampietro et al., 1997). Acute myeloid leukemia is a common complication. Some patients whose clinical condition resembles the VACTERL1 association may, in fact, have FA, and tests for chromosome breakage (see later discussion) can enable the distinction to be made (Faivre et al., 2005). Chromosomes show a range of abnormalities, including an increase in chromosome breakage, both spontaneously and upon exposure to DNA cross-linking agents (Fig. 21–1). There is little or no hypersensitivity to radiation damage. The increase in chromosome breakage after exposure of cells to a cross-linking agent such as diepoxybutane (DEB) provides, when it is observed, a reliable diagnostic test (Esmer et al., 2004; Castella et al., 2011). As Joenje et al. (1998) note, most cytogenetic laboratories will see a case of true FA only very infrequently, and it may be difficult to maintain technical expertise in the practice of clastogen-challenge test protocols. Thus, a negative result might not absolutely exclude the diagnosis. Another reason for a misleading negative result is in vivo “correction” of the functional defect in blood-forming tissue by intragenic homologous recombination, with proliferation of the corrected stem cell population. Joenje et al. refer to patients with typical FA who converted from a positive test result on blood sampling to apparent falsenegative over a period of years.2 Skin fibroblasts maintain the clastogen-sensitive phenotype, and diagnosis following fibroblast study should be reliable.
1 Vertebral, anal, cardiac, tracheo-esophageal, renal, limb. 2 This reversion to a normal cell line may work as a natural “self-treatment,” whereby the normal marrow clone arising could have a proliferative advantage and ameliorate the disease state (Gross et al., 2002).
342
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
(a)
(b) FIGURE 21–1 Metaphase from (a) a control and (b) a patient with Fanconi anemia after exposure to diepoxybutane. Note the high level of chromatid breakage in the patient metaphase. One chromatid break is indicated (straight arrow), and a quadriradial figure is shown (curved arrow).
There is genetic heterogeneity in FA, with at least 12 loci identified, mostly listed sequentially as FANCA, FANCB, and so on, to FANCP. The gene products from these different loci contribute to the control of cellular DNA repair (Kennedy and D’Andrea, 2005; Kim et al., 2011). One of the less common of these genes is the breast cancer susceptibility gene FANCD1,
better known as BRCA2; biallelic mutation leads to a particularly severe form of FA, with a very high cancer risk (Alter et al., 2007). A risk to the FA heterozygote for cancer has long been suspected (Kennedy and D’Andrea, 2005), and this consideration weighs into the counseling, with respect to the parents and wider families. However, in recent family studies from the United States and the United Kingdom, looking, respectively, at 944 and 575 relatives of patients with FA, there was no suggestion of an increased risk for cancer overall (Berwick et al., 2007; Tischkowitz et al., 2008). The specific genotypes were not assessed in the U.K. study, but the majority were likely to have been due to the FANCA genotype, and probably none due to FANCD1 (BRCA2). Three rare genotypes, FANCC, FANCJ, and FANCN, may convey a small increased risk, but confined to cancer of the breast. As for FANCD1 (BRCA2), Alter et al. (2007) observed an inconsistent association with cancer in carriers and concluded that some, but not all FA-associated mutations at this locus convey a cancer risk to the heterozygotes in the family—a conclusion that leads these authors to comment that “counseling those individuals is particularly difficult at present.” Prenatal diagnosis by mutation detection will be possible in those cases with a known mutation. Preimplantation diagnosis has been successfully applied, not only to select an unaffected embryo but also one with the same HLA typing, in order to enable blood stem cell donation to a preexisting affected sibling, an approach not without controversy (Verlinsky et al., 2001b). Otherwise, DEBinduced chromosome breakage in amniotic fluid or chorionic villus cells should provide a satisfactory approach (Auerbach et al., 1986). We have seen a case in which, at routine fetal ultrasonography, upper limb defects were identified, and the couple chose to terminate the pregnancy; subsequent analysis of fetal tissue showed the characteristic cytogenetics of FA. This same cytogenetic testing is being offered in subsequent pregnancies. Merrill et al. (2005) report somewhat similar experiences, although they were able to offer specific testing for a specific mutation enriched in the Jewish population, following ultrasound suspicions of FA.
Bloom Syndrome Bloom syndrome (BS) is a rare disorder that has its highest prevalence in Ashkenazi Jews, but it occurs Chromosome Instability Syndromes •
343
in many other ethnic groups. It is characterized clinically by proportionate short stature, a characteristic facies, sun-sensitive skin rash, immunodeficiency, and a marked susceptibility to cancer (German, 1993). Infertility seems to be invariable in the male; females have difficulty conceiving, but a few have given birth (Martin et al., 1994). The Bloom gene, BLM, was originally mapped to 15q25-qter by the elegant approach of determining the region of isodisomy in a child with BS and concomitant PraderWilli syndrome due to uniparental disomy 15 (Woodage et al., 1994). BLM codes for a recQ DNA helicase that monitors DNA integrity during S phase (German and Ellis, 2011). (Another member of this gene family is the basis of Rothmund-Thomson syndrome.) The diagnostic cytogenetic finding in BS is a markedly increased level of spontaneous sister chromatid exchange (SCE). The normal is 6 to 10 exchanges per cell; in BS, it is more than 50 per cell (Fig. 21–2), although some normal cells may be present in BS patients.3 The other cytogenetic abnormality is an increased incidence of spontaneous chromatid aberrations, giving the classic symmetrical quadriradial configuration. Intriguingly, this effect can manifest in the haploid state, with the heterozygous male producing an excess of sperm with chromosome breaks and rearrangements (Martin et al., 1994c). Prenatal diagnosis may be based upon observation of increased sister chromatid exchanges in chorionic villus cells (Howell and Davies, 1994). Specific mutation analysis would be applicable if the family mutations were known; a Bloom mutation register is maintained (German et al., 2007). For the affected woman’s reproductive outlook (in those few surviving to adulthood), the standard mendelian advice, with consideration of the likelihood of the spouse being heterozygous, applies (Chisholm et al., 2001). As noted earlier, the male is infertile.
(a)
(b) FIGURE 21–2 Metaphase from (a) a control and (b) a patient with Bloom syndrome, showing very high sister chromatid exchange (SCE) in the latter. Three points of SCE are indicated (arrows) on the control metaphase.
Ataxia-Telangiectasia Ataxia-telangiectasia (AT) is the archetype of a group in which the basic pathogenetic process is a failure in one of the monitoring and repair systems that keep watch for DNA damage. The group includes AT itself, and Nijmegen breakage syndrome (below),
and both exhibit chromosome instability. The genes for AT and Nijmegen breakage syndrome encode proteins that are part of a complex sensing abnormal DNA structures and monitoring postreplication DNA repair (Michelson and Weinert, 2000).
3 Interestingly, the normal cells may be due to a “correcting” genetic event occurring in a bone marrow cell, and which then leads to a heterozygous cell line having a normal in vitro phenotype. The correcting event may be either a somatic recombination between the two sites of BLM mutation in the homologs in the BS individual with compound heterozygosity, or a back mutation in a homozygote (Ellis et al., 2001).
344
•
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
The clinical presentation of AT is as a brain/ immune/cancer syndrome. It is characterized by cerebellar ataxia and oculomotor apraxia (difficulty in performing voluntary eye movements), oculocutaneous telangiectasia, immunodeficiency, and increased cancer predisposition. The cytogenetic hallmarks of AT include frequent nonrandom rearrangements of chromosomes 7, 14, and occasionally X, in T-lymphocytes; nonspecific chromosome breakage in fibroblasts; and normal chromosomes in bone marrow. The breakpoints in the lymphocyte rearrangements are at 7p14, 7q35, 14q12, and 14q32, involving the T-cell receptor and immunoglobulin heavy chain genes. Clones with rearrangements may be harbingers of a T-cell malignancy, and these clones evolve as the disease progresses. Breakage is exacerbated, in vitro, by exposure of cells to ionizing radiation and radiomimetic chemicals such as bleomycin (Kojis et al., 1991). Most ATM mutations are null, but missense and splicing mutations that allow a limited amount of functional product to be produced may lead to milder clinical and cytogenetic phenotypes. Some of these “milder” mutations may, on the other hand, promote an increased cancer risk, including breast cancer in the female heterozygote (Chenevix-Trench et al., 2002). Prenatal diagnosis of classic AT can be approached cytogenetically on amniocyte analysis, or it may be achievable using direct mutation analysis of the ATM gene on chorionic villus tissue (Chessa et al., 1999). Preimplantation genetic diagnosis may be successful (Hellani et al., 2002).
Nijmegen Breakage Syndrome This is another brain/immune/cancer syndrome, and it is rare indeed. The clinical picture includes microcephaly with brain dysgenesis, immune deficiency, and risk for lymphoreticular malignancy. It shares with AT certain cytogenetic features (preferential involvement of chromosomes 7 and 14 in rearrangements) and radiation hypersensitivity (Antoccia et al., 2006). The causative gene, called NBS1, interacts with the ATM gene, noted earlier. A founder mutation, 657del5, is common amongst the Slavic population, and most patients are 657del5 homozygotes (Seemanová et al., 2006). Prenatal diagnosis is preferably achieved by specific mutational analysis.
Roberts Syndrome Roberts syndrome (RS) is a syndrome of craniofacial abnormalities and limb defects that are often
severe, and the archetype of the “cohesinopathies”. The phenotype is so very distinctive that is unsurprising that case reports go back some centuries, the first appearing in 1672 (a “Portrait d’un enfant monstre”), well before Roberts’ description from 1919 (Bates, 2003; Kompanje, 2009). Intellect is normal. Most affected individuals (about 80%) exhibit a chromosomal phenomenon known as premature chromatid separation (PCS), sometimes described as “tram-tracking” or “railroad track appearance,” and also referred to as “heterochromatin repulsion,” as the sister chromatids bulge away from each other. The gene is ESCO2 (Vega et al., 2010), and its product enables proper disposition of the chromatids. In its absence, there is an abnormality of sister chromatid apposition around the centromeres, particularly noticeable for those chromosomes with large blocks of heterochromatin (Fig. 21–3). It is best seen in plain stained or C-banded chromosomes; G-banding obscures the phenomenon (Van Den Berg and Francke, 1993). In this particular instance, classical cytogenetics is the more powerful diagnostic tool, and it may enable recognition of an atypical case; microarray would miss the abnormality (Gerkes et al., 2010). Prenatal diagnosis based upon the presence or absence of PCS at chorionic villus sampling or amniocentesis, and abnormality or normality of limbs on first-trimester fetal ultrasonography, should be valid in at least the majority. It would be prudent to follow up an interpretation of normality at secondtrimester ultrasonography. Molecular testing can be applied when the specific mutation is known (Schulz et al., 2008).
Cornelia de Lange Syndrome The clinical phenotype of CdeLS is, in the classic case, very distinctive. Three causative genes are known: NIPBL (the most frequently seen), SMC1A, and SMC3. In contrast to the recessive basis of the foregoing conditions, the inheritance with NIPBL and SMC1A is typically de novo heterozygous mutation, although rare familial transmission is recorded (Russell et al., 2001; Krantz et al., 2004). SMC3 is X-linked (Musio et al., 2006). Other genes may exist. The cytogenetic phenotype is premature chromatid separation (PCS), and this condition is another cohesinopathy. Testing for PCS may be a useful adjunctive test in the diagnosis of CdLS, if mutation analysis is not available, or is negative (Kaur et al., 2005). Chromosome Instability Syndromes •
345
(a)
(c)
(b)
(d)
FIGURE 21–3 Unusual appearance of the chromosomes in Roberts syndrome: puffing at the centromeres (a and b); a C-banded preparation showing separation of the heterochromatic segments (c) is compared with a C-banded preparation from a control showing the normal centromere appearance (d). (From N. P. Mann et al., 1982, Roberts syndrome: clinical and cytogenetic aspects, Journal of Medical Genetics 19:116–119. Reproduced with the permission of the British Medical Association.)
VE RY RA RE SY N D ROM ES
ICF (Immunodeficiency, Centromeric Instability, Facial Anomalies) Syndrome. The ICF syndrome is characterized by immunodeficiency, an unusual facies, and growth and developmental retardation; and a most remarkable tendency of chromosomes nos. 1, 9, and 16 to form “windmill” multiradials by interchange within heterochromatic regions (Fig. 21–4). This instability of the pericentromeric heterochromatin reflects hypomethylation of satellites 346
•
II and III, which are important components of its structure. Hagleitner et al. (2008) document the variability of the phenotypic range. The phenotype, physical and cytogenetic, can be considered to be secondary to a failure of methylation. Most cases are due to mutation in the DNA methyltransferase 3B gene, although locus heterogeneity is presumed to exist. Variegated Aneuploidy with Premature Centromere Separation. The core phenotype of
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
Seckel Syndrome. At least three loci may exist, SCKL1–3. In a group of five patients with this syndrome of growth retardation, microcephaly and characteristic facies, Bobabilla-Morales et al. (2003) demonstrated excessive chromosomal breakage, although not an excess of sister chromatid exchanges. Casper et al. (2004) discovered, in patients with SCKL1 (due to the ATR gene, which interacts with ATM), that the breaks cluster at common fragile sites. Some SCKL forms do not display chromosome instability.
FIGURE 21–4 A “windmill” or “starburst” multiradial chromosome 1 in the ICF syndrome. (From J. R. Sawyer et al., 1995, Chromosome instability in ICF syndrome: formation of micronuclei from multibranched chromosomes 1 demonstrated by fluorescence in situ hybridization, American Journal of Medical Genetics 56:203–209. Courtesy J. R. Sawyer; reproduced with the permission of Wiley-Liss.)
this recessively inherited syndrome comprises microcephaly with functional neurological abnormality, growth retardation, and susceptibility to childhood malignancy, with most of the lymphocytes and about half of skin fibroblasts showing premature chromatid separation. Many cells are aneuploid, with trisomies, double trisomies, and monosomies, with almost every chromosome represented, and sometimes referred to as “mosaic variegated aneuploidy” (Bohers et al., 2008; García-Castillo et al., 2008). The underlying defect in the cell cycle involves one of the checkpoint proteins (BUB1B) that control progression through the mitotic process, maintaining an alert for chromosome malsegregation. The BUB1B heterozygote may display the tendency in a proportion of lymphocytes, and some mitotic cells may present the striking observation of a 92-chromosome count. Prenatal diagnosis has been reported, based on conventional cytogenetics, the abnormalities being very obvious (Plaja et al., 2003; Chen et al., 2004c).
Syndromes Reported in Only One Family (A Few Examples). • Ishikawa et al. (2000) reported a single family with a dominantly inherited chromosome instability syndrome. The major clinical observations are mild to moderate mental retardation, depression, and a spastic ataxia, with striking abnormalities of cerebral white matter and the basal ganglia, and an atrophic spinal cord. All three affected individuals having a cytogenetic analysis showed a low frequency of a t(7;14), with a common 14q11.2 breakpoint in each, and a hypersensitivity to radiation and radiomimetic drugs. • A unique Austrian family appears to present a sex-limited chromosome breakage syndrome with ovarian failure (Duba et al., 1997). The index case had presented with primary hypogonadism, and karyotyping showed a high proportion of cells with breaks, acentric fragments, triradial rearrangements, and dicentric chromosomes. Two healthy brothers had essentially the same chromosome findings. The cytogenetic picture most closely resembled that of Fanconi anemia, and the three siblings also demonstrated an elevation in α-fetoprotein, which is a feature of AT. Lespinasse et al. (2005) report a similar case, but in this instance, a sister and a brother were both infertile, and the α-fetoprotein normal. • Bakhshi et al. (2006) describe a 17-year-old boy with growth retardation and dysmorphic facies, with mitomycin-sensitive chromosomal breakage, who developed a B-cell lymphoma; they proposed this as a new syndrome, distinct from FA. • A severely growth-retarded and microcephalic teenager showed both chromosomal breakage and premature chromatid separation, and represents a further cohesinopathy, named Chromosome Instability Syndromes •
347
Warsaw breakage syndrome for the city of his residence (van der Lelij et al., 2010). The causative gene is DDX11, having some sequence similarity to the gene for Fanconi Anemia type J, and coding for a helicase. Inheritance is autosomal recessive, although there is a hint the heterozygote may have an increased cancer risk (and see FANCJ mentioned earlier). • While not a breakage syndrome, the novel disorder reported by Neitzel et al. (2002)
348
•
deserves a mention. The cytogenetic observation is that of chromosomes prematurely entering mitosis: in metaphase lymphocytes (without colcemid exposure) and fibroblasts, an excess of cells showed chromosomes appearing to be in prophase. The two affected children were severely retarded in growth and mental development. The condition is presumed autosomal recessive.
N O R M A L PA R E N TS W I T H A C H R O M O S O M A L LY A B N O R M A L C H I L D
PART FIVE DISORDERS ASSOCIATED WITH ABERRANT GENOMIC IMPRINTING
This page intentionally left blank
22 uniparental disomy and disorders of imprinting
UNIPARENTAL DISOMY IS A FASCINATING and important pathogenetic mechanism, albeit that it is the basis of only a small number of well-defined clinical conditions. At the outset, we may list these seven major syndromes: Prader-Willi syndrome Angelman syndrome Beckwith-Wiedemann syndrome Silver-Russell syndrome Transient neonatal diabetes Maternal uniparental disomy 14 (Temple syndrome) Paternal uniparental disomy 14 Prader-Willi syndrome, Angelman syndrome, and Beckwith-Wiedemann syndrome can be due to
other genetic causes in addition to uniparental disomy (UPD1), and for convenience we include a discussion of these other causes in this chapter. As well as the aforementioned seven conditions, certain other UPDs can be the cause of abnormality. These may manifest, in various combinations, the following traits: intrauterine and postnatal growth retardation, intellectual deficit, congenital malformations, and dysmorphic features. In the small print is first, pseudohypoparathyroidism type 1B, due to upd(20)pat, and second (although this may come to demand a larger-print awareness), the maternal hypomethylation syndrome, which has a particular association with in vitro fertilization (IVF) conceptions (Amor and Halliday, 2008). In a category by itself, UPD can be the cause of homozygosity for an autosomal recessive gene. The foregoing notwithstanding,
1 As a general rule, the abbreviation is in uppercase (UPD, UPHD, UPID) when making broad reference to the concept of uniparental disomy, and in lowercase (upd, uphd, upid), according to the rules of cytogenetic nomenclature, when attention is more focused upon a specific case.
•
351
however, the fact remains that most UPDs appear to be without any phenotypic consequence, and a number of syndromes that had seemed fair candidates turned out not to be due to UPD (Kotzot, 2002). A distinction is to be made between UPD where both chromosomes are identical (uniparental isodisomy, UPID) and where they are different (uniparental heterodisomy, UPHD) (Fig. 22–1). UPD is normally demonstrable only at the molecular level: typically, although not invariably, the UPD pair of chromosomes are cytogenetically normal, and the karyotype appears normal, 46,XX or 46,XY. The pattern of polymorphic DNA markers shows that both chromosomes have the same haplotype as just one of the chromosomes from one of the parents (isodisomy); or the two chromosomes have the same haplotypes as the chromosome pair from one of the parents (heterodisomy). For example, the chromosome 1 haplotypes from parents and child set out in Figure 22–1b show that the child has two identical copies of one of the father’s chromosomes: thus, paternal uniparental isodisomy. This UPD had been discovered fortuitously, when the child was investigated for a clinical diagnosis of congenital insensitivity to pain, an autosomal recessive disorder (Miura et al., 2000). He proved to be homozygous for a mutation in the appropriate gene (TRKA, located at 1q21-q22), and his father carried the mutation, but his mother did not. This scenario—a child with a recessive disorder for which only one parent is heterozygous—is commonly the circumstance behind the discovery of UPIDs that would
(a)
(b)
FIGURE 22–1 (a) The distinction between uniparental heterodisomy and uniparental isodisomy. The four parental homologs are shown in different patterns. In the child with heterodisomy, the two homologs are different. In isodisomy, they are identical. Meiotic crossing-over can lead to segmental iso/heterodisomy, and the pattern can reveal whether the initial nondisjunction had been at meiosis I or II (see text). (b) The molecular picture of a child with paternal uniparental isodisomy 1. The markers run from D1S468 at the top of chromosome 1 down to D1S2836 at the bottom. Both the child’s chromosome
352
•
1 haplotypes are the same, and the same as one of his father’s no. 1 chromosomes. He has no chromosome 1 from his mother. (The arrow points to the position of the TRKA locus. Homozygosity for an abnormal TRKA allele was the cause of his having the recessive condition congenital insensitivity to pain, which had led to his ascertainment.) (From Y. Miura et al., 2000, Complete paternal uniparental isodisomy for chromosome 1 revealed by mutation analyses of the TRKA (NTRK1) gene encoding a receptor tyrosine kinase for nerve growth factor in a patient with congenital insensitivity to pain with anhidrosis, Human Genetics 107:205–209. Courtesy Y. Indo, and with the permission of Springer-Verlag.)
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
otherwise have been without clinical effect. The other typical route to recognition of harmless UPDs is an incidental discovery in the course of polymorphic DNA marker analysis being done for other reasons. The state of iso- or heterodisomy can allow an inference as to the site of the initial chromosomal error. Isodisomy typically reflects a meiosis II nondisjunction or a mitotic error, whereas heterodisomy is due to nondisjunction at meiosis I. Partial heterodisomy and partial isodisomy can coexist for the same chromosome pair. For example, a crossover at meiosis I in, say, the distal long arm, followed by meiosis I nondisjunction, could lead to a disomic gamete isodisomic for distal long arm, and heterodisomic for proximal long arm (Fig. 22–1a, lower right). If the nondisjunction were at meiosis II, the isodisomy and heterodisomy would be the other way around, involving the proximal and distal segments, respectively (Fig. 22–1a, lower left). E PI G EN ETICS A N D IM PRI NT I N G
The definition of epigenetic has been evolving (Berger et al., 2009). A core consideration is that a phenotype may differ according to whether a DNA sequence is active, or inactive, but the DNA sequence itself remains unchanged. Our focus is on the activity or nonactivity of a gene (or chromosomal segment) according to the parental origin of the chromosome upon which the gene (or segment) is located. Thus, a chromosomal segment can receive an “epigenetic mark”—or is “imprinted”—as it is transmitted from parent to child, depending upon whether it is the mother or the father who has contributed that chromosomal segment, and this determines whether this segment will be genetically active or not active (“silent”). This is spoken of as a “parent of origin” effect. The major physical basis of this epigenetic effect is due to methylation of the DNA (that is, a methyl group attached to cytosine bases), modification of the histone scaffolding of chromatin, and the actions of noncoding RNAs, which severally or separately can then prevent the expression pattern of the relevant gene(s). There are certain chromosome segments (in sum, only a small fraction of the whole genome) that are subject to imprinting.2
A little counterintuitively, imprinting refers to nonactivity: an imprinted chromosome segment is silenced, while the nonimprinted chromosome segment is the active one. In the normal setting, with biparental inheritance, imprintable segments (or loci) function monoallelically. That is to say, it is only the segment of maternal origin, or only the segment of paternal origin, as the case may be, which is genetically active. But if both segments originate from one parent, there will be either double the amount (biallelic) of expression, or no (nulliallelic) expression, according to the gender of the contributing parent. (Some imprinting is tissue specific, in which case, the aberrant expression is confined to that tissue.) It is this functional imbalance that is the root cause of the phenotypic effect in the UPD syndromes. If a chromosome is not subject to imprinting, UPD does not of itself cause abnormality, other things being equal. The only other factor due to UPD, and specifically UPID, which can lead to defect, is homozygosity for a recessive mutation (“isozygosity”), as noted earlier.
Uniparental Disomy for a Complete Chromosome In UPD for a complete and intact chromosome, both members of a homologous pair come from the one parent. Four routes to lead to this state are the following (and see Fig. 22–2): • Gametic complementation • Trisomic rescue • Monosomic rescue • Mitotic error Gametic complementation is mentioned first, as the simplest and classic example, but in truth it must hardly ever be that UPD is the consequence of a meiotic error happening coincidentally in both parents (Park et al., 1998; Shaffer et al., 1998). Trisomy “rescue” or “correction”3 is the mechanism behind most UPD. The cause of the trisomy is a typical meiotic nondisjunction that happened in one of the two conceiving gametes. The rescue process takes place in a cell of the trisomic conceptus
2 The subject is, however, becoming more complicated, with the recognition that some nonimprintable genes may normally function monoallelically, and that different parent-of-origin epigenetic influences upon many loci can operate in different anatomical brain regions during development (Gimelbrant et al., 2007; Gregg et al., 2010).
Uniparental Disomy and Disorders of Imprinting
•
353
FIGURE 22–2 Mechanisms whereby complete uniparental disomy (UPD) may be generated. (a) Gametic complementation, with one parent producing a disomic gamete, and the other a nullisomic gamete. (b) Meiotic nondisjunction in one parent to produce a disomic gamete, with a trisomic conceptus following fertilization, and subsequent mitotic loss of the homolog from the other parent. This is uniparental heterodisomy, from the parent in whom the nondisjunction had taken place. (c) Meiotic nondisjunction in one parent to produce a nullisomic gamete, with monosomic conceptus following fertilization, and subsequent mitotic reduplication of the homolog from the other parent. This is uniparental isodisomy, from the parent who had contributed the normal gamete. The reduplication may produce a free homolog or an isochromosome. (d) Two sequential mitotic errors. *Since most meiotic nondisjunction occurs in maternal gametogenesis, these asterisked gametes can be imagined to be oöcytes, with UPD(mat) and UPD(pat) resulting accordingly.
at a very early postzygotic stage (possibly even in the zygote), with one of the trisomic chromosomes being discarded, perhaps due to anaphase lag. This enables a cell line within the conceptus to be restored to disomy, but in order to cause the subsequent UPD it is the “wrong” chromosome that is eliminated— that is, purely by chance, the discarded chromosome happens to be the one that came from the normal gamete, and so the remaining two are from the same parent. These two chromosomes will comprise one of each of the homologs of that parent: thus, uniparental heterodisomy. This would be expected to happen, by chance, in one-third of rescues, biparental inheritance being maintained in the other two-thirds (close to these ratios was observed in a large study of UPD16; Yong et al., 2002). The 46-chromosome cell with UPD that results from this process may be the progenitor of the cells which produce the inner cell mass, which in turn gives rise to the embryo. Any remaining trisomic cells may go on to form the placenta, leading to confined placental mosaicism, or they may also contribute to the inner cell mass, leading to trisomy/disomy mosaicism of the embryo. Thus, the phenotypes in some UPD states are complicated by the additional effects of compromised placental function due to trisomy, and/or of fetal trisomy mosaicism. Monosomic rescue also comes into play following a nondisjunctional event. If a nullisomic gamete is generated at meiosis, then the conceptus will be monosomic (assuming a normal gamete from the other parent). Mitotic correction then takes place, and this is achieved by replication of the single, normal, homolog received from the other parent. In this case, the UPD will be an isodisomy. The fourth possibility is a mitotic error in an initially normal conception, leading to either trisomy or monosomy. In the case of a trisomy, this is followed shortly thereafter by loss, in this cell line, of the nonreplicated trisomic chromosome. In the case of a mitotic nondisjunction resulting in monosomy, the remaining homolog is then duplicated. In both cases, the UPD is isodisomic. Note that each of these four scenarios requires there to be two separate abnormal events, occurring either simultaneously (the first scenario) or sequentially (the latter three). These errors can be both meiotic (the first), meiotic followed by mitotic
(second and third), or both mitotic (the fourth). In whichever case, the original abnormality will practically always have been a sporadic event, with no discernible increased risk of recurrence due to having had one affected child; and indeed, to our awareness, as yet not one instance is known of a recurrence of UPD in the setting of normal parental karyotypes. One risk factor is known, and this is increasing maternal age. The link here is that meiotic nondisjunction, the root cause of most UPD, is more prevalent in women of older childbearing age. The meiotic errors noted earlier as leading to trisomic rescue and monosomic rescue are typically of maternal origin. Ginsburg et al. (2000) have shown that maternal age is higher in the subset of patients with Prader-Willi, Angelman, and Russell-Silver syndromes due to UPD, compared to those due to other causes. A reduced level of recombination is seen in UPD 15 (Robinson et al., 1998), an observation also made in the classic disorder with a maternal age association, namely Down syndrome. It is worth noting that paternal UPD also has a maternal age effect, which seeming contradictory statement can be appreciated upon considering the mechanism of monosomic rescue after mostly maternal nondisjunction, this being the usual initiating cause of UPDpat. Rare mechanisms to generate complete UPD include the following: • Correction of interchange trisomy • Correction of interchange monosomy • Isochromosome formation • Correction of imbalance due to small marker chromosome If one parent carries a reciprocal translocation, asymmetric segregation of the chromosomes may lead to an interchange trisomy (p. 89) at conception, in which the translocation chromosomes, plus one of the normal homologs, are transmitted. Postzygotic correction by the loss of one homolog restores disomy, but if it is the other parent’s chromosome that is lost, UPD is the consequence. Or, if a nullisomic gamete meets a normal gamete (interchange monosomy), the normal gamete may replicate the homolog in question, to restore disomy (just as in monosomy rescue, mentioned earlier).
3 It might be more accurate to speak of a “failed rescue,” or better a “foiled rescue,” since the end result is an unfortunate one. Or “mistaken correction.”
Uniparental Disomy and Disorders of Imprinting
•
355
Kotzot (2001) recorded 22 examples of UPD associated with a reciprocal or Robertsonian translocation, involving UPDs for chromosomes 7, 13, 14, 15, and 16. In the case of parent with a Robertsonian translocation, the most common mechanism leading to UPD is a trisomy rescue after nondisjunction. A monosomic acrocentric chromosome, after nondisjunction from a Robertsonian translocation parent and fertilization with a normal gamete, could replicate as an isochromosome in a monosomy rescue (Berend et al., 2000a; McGowan et al., 2002). Complementary isochromosomes (p. 157), of which scarcely a double-digit number have ever been described, can even allow the circumstance of “contraposed UPD”: that is, there may be UPD of the p arm from one parent, and UPD of the q arm from the other. Finally, in the setting of a supernumerary small marker chromosome (SMC), there may be a coexisting UPD for the same chromosome from which the SMC was derived ( James et al., 1995; Liehr, 2010).
Segmental Uniparental Disomy Segmental UPD may arise as the consequence of a postzygotic somatic recombination, between the maternal and paternal homolog (Fig. 22–3), and in that case it will necessarily be an isodisomy (Kotzot, 2008a). An assessment of “long contiguous stretches of homozygosity” may prove a useful means to demonstrate the state (Papenhausen et al., 2011). The UPD segment lies distally, the rest of the chromosome having a normal biparental disomy. The classical karyotype is normal. An alternative mechanism is the following sequence: meiotic nondisjunction producing a disomic gamete, a trisomic conception, a mitotic crossing-over between a maternal and a paternal chromatid, and finally loss of one of the chromosomes that had come with the disomic gamete. UPD can have an effect if the particular chromosomal segment incorporates loci subject to imprinting. If the recombination occurs in a cell
FIGURE 22–3 A mechanism whereby segmental uniparental (iso)disomy may be generated. In one cell of the early conceptus, the paternal and maternal homologs of a chromosome pair (a) undergo somatic recombination between the short arms (b and c). Segregation at mitosis (d) produces daughter cells with segmental uniparental disomy (UPD): in one (e), the short arm distal segments of both chromosomes are now of paternal origin, and in the other (f), they are both of maternal origin. These cells can then be the source of segmentally UPD tissue in a part of the conceptus.9 9 The same mechanism of postzygotic recombination may suggest itself in the setting of somatic mosaicism for a mendelian condition. Happle and König (1999) discuss the case of a boy with a rare skin condition (epidermolytic hyperkeratosis of Brocq) affecting most of his body, but with some areas of more severely affected skin, and some areas which were healthy. Imagining that the Brocq locus might be in the distal short arm of the chromosomes depicted in Figure 20–3 and with the black chromosome having the mutant allele, the typically affected skin would have the genotype represented in (a), the more severely affected skin would be in (f), and the normal skin in (e).
356
•
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
after the formation of the inner cell mass (which gives rise to the embryo), the segmental UPD will involve only some cells; in other words, there is mosaic segmental UPD. Beckwith-Wiedemann syndrome, Russell-Silver syndrome, UPDs for chromosome 14, and transient neonatal diabetes mellitus are conditions in which segmental UPD may apply. If a segment subject to UPID, due to one of the aforementioned scenarios, contains a recessive mutation, the disease related to that mutation will result. An example is given in Wattanasirichaigoon et al. (2008), who describe a woman heterozygous for the common Southeast Asian α-globin deletion on chromosome 16p, and who had a fetus with hemoglobin Bart’s hydrops, a lethal disease. The likely scenario is that the ovum contained two copies of chromosome 16, which were isodisomic for distal 16p due to meiosis I recombination. Fertilization led to trisomy 16, but with the paternal homolog then lost by “rescue.” Thus, the resultant fetal genotype was homozygosity for the α-globin deletion. A partial trisomy might have different abnormal phenotypic effects according to the parental origin of the duplicated segment, if that segment is subject to imprinting. Trisomy for distal 14q provides an example. A similar picture of dysmorphology and psychomotor deficit is seen in either paternally or maternally originating 14q trisomy. But low birth weight, sometimes less than 2000 grams for a full-term baby, is a specific observation when the duplicated 14q segment comes from the mother (Georgiades et al., 1998). A classic example is the dup15q13, described in detail on p. 329: inherited from the father, there is typically no phenotypic consequence, but when the duplication is transmitted maternally, the child develops autism. Aberrant Imprinting in a Biparental Setting. A chromosomal segment that is normally imprinted (thus, inactive) may lose its imprint and become active. This is “relaxation” (or inappropriate erasure) of the imprint effect, and it may be termed an “epimutation”; to reemphasize the point, the DNA sequence remains unchanged. Consider BeckwithWiedemann syndrome (BWS). In some BWS with normal biparental inheritance of chromosome 11, the IGF-2 (insulin-like growth factor 2) and KCNQ1OT1 loci on distal 11p show biallelic expression; normally, only the paternal alleles should be functional. This overexpression of genes contributes to the overgrowth that is characteristic of the
syndrome (as discussed in more detail later). An iatrogenic cause of aberrant imprinting may relate to pregnancy following assisted reproductive technology; aspects of the process of artificial ovulation stimulation, or of the embryo’s environment in vitro, may disturb DNA methylation (Kagami et al., 2007; Amor and Halliday, 2008; Khoureiry et al., 2008; Marques et al., 2008; Katari et al., 2009).
Uniparental Disomy Phenotypes Uniparental disomy has been observed for every chromosome except for 19 (Liehr, 2010). For most chromosomes, there is no apparent phenotypic consequence. For others, there may be, and we list below some of the proposed syndromes of UPD. The reader seeking more detail is referred to Engel and Antonarakis (2002), Kotzot (2008a), and Amor and Halliday (2008). Some conditions in which UPD had been thought of as a fair possibility have turned out not to be: Cornelia de Lange syndrome and Sotos syndrome, for example. In the case of UPD arising from incomplete trisomic rescue, additional factors of trisomy of the placenta, and/or a residual low-level trisomy of the fetus, may also contribute to the eventual phenotype. De Pater et al. (1997) note that a fetal trisomic cell line may not be detected unless the possibility of mosaicism is painstakingly pursued, and Benn (1998) uses the expression “occult mosaicism” to denote an unprovable suspicion. Because mosaicism can never be completely excluded, and neither can homozygosity for an unknown recessive mutation, one should generally incline in the direction of accepting that there is an absence of any UPD effect, when instances are known both of normal and of abnormal phenotypes, or when the observed abnormalities are inconsistent (Kotzot, 1999). The abnormal phenotypes will more likely be due to non-UPD mechanisms. Certain clinical groups might be considered as candidates to harbor cases of UPD. Intrauterine growth retardation (IUGR) is an obvious category. Moore et al. (1997) addressed the question in a study of a cohort of 35 severely affected babies with a 46,N karyotype. Two instances of UPD (5% of the total) were identified, both with upd(16), and in each a coexisting placental 47,+16/46 mosaicism was shown. Neither baby survived. Kotzot et al. (2000a), checking chromosomes 2, 6, 14, 16, 20, and 22, found no instances of UPD in a series of 23 cases of IUGR, using a broader definition of birth weight and/or length below the 10th centile (10 of Uniparental Disomy and Disorders of Imprinting
•
357
the 23 were considered to have Silver-Russell syndrome). Eggermann et al. (2001) studied 21 patients with pre- and postnatal growth retardation, choosing chromosomes 2, 7, 9, 14, 16, and 20 for analysis, and identified one with upd(14)mat and one with upd(20)mat. Except for the upd(20)mat, which may or may not have been the cause of the child’s “minor features,” it is to be noted that only already known UPDs (for 7, 14, 15, and 16) were identified in these several surveys. “Unclassified congenital developmental defects” was the criterion for entry to the study of Ginsburg et al. (2000), comprising a cohort of 50 individuals, whose mothers had been aged 35 years or older at the time of delivery. This sort of patient is, of course, very familiar to the genetic counselor. Four turned out to have a UPD. The specific UPDs were as follows: maternal heterodisomy 14 in a woman with short stature and early puberty; paternal isodisomy 15 in a boy with previously undiagnosed (but retrospectively apparent) Angelman syndrome; upd(16) mat along with a partial 16p trisomy; and a child with, in retrospect, Silver-Russell syndrome, having a upd(7)mat. Four out of fifty (8%) may be a higher fraction than could usually be expected, and it is easy to be wise after the event that two of the cases were not really “unclassifiable.” A lower fraction, namely 0%, was observed in a larger study of 120 children with two or more malformations, developmental or growth retardation, and a normal routine karyotype (Rosenberg et al., 2001). Concerning a possible contribution to spontaneous abortion, Shaffer et al. (1998) studied 18 cases of cytogenetically normal first-trimester miscarriages, with every chromosome analyzed, but none showed UPD. In a more extensive study, Fritz et al. (2001) analyzed products of conception along with parental blood samples, which included some 77 spontaneous abortions with normal karyotypes. Of these, only two showed UPD: one case of maternal UPHD 9, and one of paternal UPID 21. Thus, from these two reports, only 2% of karyotypically normal spontaneous abortion is associated (whether or not this be causally) with UPD. We have already noted the UPD effect of reduction to homozygosity of a recessive mutation. Engel and Antonarakis (2002) list 22 reports in which this sort of mechanism has been identified, including one notable example in which a child had both cystic fibrosis and Kartagener syndrome, the loci for these two separate recessive disorders lying on chromosome 7. The most extraordinary case is that of a 358
•
couple, both normal homozygotes, whose child had maple syrup urine disease due to fresh mutation in oögenesis, with meiosis II nondisjunction then producing an isodisomic ovum (Lebo et al., 2000). We list, by individual chromosome, the UPD syndromes, or associations with normality, that are on record. In each, the additional comment can be made, as just mentioned, that UPID could lead to unmasking of a recessive disorder; and likewise that an undetected residual trisomy might contribute to a phenotype, when the UPD mechanism has been trisomy correction. Chromosome 1. Maternal UPD of chromosome 1 may have of itself no effect (provided no recessive mutations are unmasked, as exemplified in Miura et al., 2000, and illustrated in Fig. 22-1b). Field et al. (1998) made the serendipitous discovery of UPD 1 in a normal diabetic adult in the course of a genetic study of diabetes, as did Miyoshi et al. (2001) in their investigation of two normal persons with anomalous Rh blood grouping results: upd(1)mat in the former, mosaicism for paternal isodisomy 1 in the latter. Unmasking of recessive genes, rather than an effect of imprinting, may have been the basis of phenotypic abnormality in a unique case of upd(1) pat described in Chen et al. (1999c). A woman of normal intelligence had a myopathy, short stature, sterility, and deafness. In this case, there was a paternal isodisomy, with the chromosome 1 elements present in the form of two isochromosomes, i(1) (p10) and i(1)(q10). Chromosome 2. It is yet unclear whether a maternal UPD2 syndrome exists (Shaffer et al., 1997b; Wolstenholme et al., 2001b). In five patients, the recurrent observations included intrauterine and postnatal growth retardation (four of five cases), atypical bronchopulmonary dysplasia/hypoplasia (three cases), and hypospadias (two cases). Isozygosity for a recessive mutation, in this case the ABCA12 gene located at 2q34 that is the basis of severe harlequin ichthyosis, was the result of trisomic rescue in a case reported by Castiglia et al. (2009), an interpretation underpinned by the observation of nonmosaic trisomy 2 at chorionic villus sampling. Chromosome 3. There is a single case from the earlier literature (Betz et al., 1974) of a retarded girl homozygous for a rare cytogenetic polymorphism, carried by only one parent, which might possibly be an example of upd(3).
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
Chromosome 4. UPD4mat, isodisomic or heterodisomic, may be without a phenotype per se, the inconsistent abnormalities in the cases studied (three iso-, one heterodisomic) conceivably having some other cause (Kuchinka et al., 2001; Middleton et al., 2006). Cottrell et al. (2011) report a case of (autosomal recessive) limb girdle muscular dystrophy type E, for which the suggested sequence of events was as follows: mother heterozygous for recessive mutation; advanced maternal age; aberrant recombination between chromosome 4 homologs at maternal meiosis; meiotic nondisjunction; trisomy 4 conception; trisomy rescue; maternal UPD 4; isozygosity of the causative gene. Chromosome 5. Paternal UPD5 may be of no effect (Engel and Antonarakis, 2002). Chromosome 6. The defining feature of transient neonatal diabetes mellitus (TNDM) is hyperglycemia requiring treatment with insulin, with a gradual resolution to normal glucose metabolism in the first few months of life, although with a risk subsequently for non-insulin-dependent diabetes in adult life. In at least a substantial fraction of cases, TNDM is due to upd(6)pat, with 6q22-q24 being the crucial region. A methylation defect within the critical region, or 6q22–q24 duplication of paternal origin, this latter accounting for all familial cases, are other routes whereby TNDM may arise (Temple and Shield, 2002). One example due to a familial insertion involving the segment 6q22-q23 is mentioned on p. 237. Parker et al. (2006) describe a child with congenital adrenal hyperplasia (the 21-hydroxylase gene being on chromosome 6) and Klinefelter syndrome, 48,XXY,+mar(6), with maternal isodisomy for both chromosomes 6 and X. “Correction” of fetal trisomy 6 was the probable basis of upd(6) mat identified by Cockwell et al. (2006) in a case of fetal death in utero. But upd(6)pat can also be without apparent effect, as witness an otherwise normal girl with thalassemia whose family was being studied to find a donor for marrow transplantation, and who turned out to have paternal UPID 6 (Bittencourt et al., 1997). Chromosome 7. Silver-Russell syndrome (SRS) has as its major feature intra-uterine and postnatal growth retardation, often with associated limb asymmetry. Genetic causes include maternal UPD 7 (about 10%), and 11p15 epimutation and structural 11p aberrations (see section on “Chromosome 11”),
with clinical and endocrine observations differing somewhat in each type (Binder et al., 2008; Wakeling et al., 2010). There is a maternal age association: very few SRS children born to mothers under age 35 have UPD 7, but around half of those born to mothers 35 or over are due to upd(7)mat, and this may reflect a maternal meiosis I error as the underlying cause (Ginsburg et al., 2000). One case is recorded of SRS in the setting of a maternal reciprocal translocation involving chromosome 7: the conception was probably an interchange trisomy, 47,t(7;16) (q21;q24),+7, with subsequent loss of the paternal chromosome 7 producing the balanced state, but with a maternal UPHD 7 (Dupont et al., 2002). As for paternal UPID 7, only two cases have been discovered (Engel and Antonarakis, 2002). One was a woman of normal linear growth, and a normal intellect, and it was only because she had a recessive condition with its locus on chromosome 7 (congenital chloride diarrhea) that she had been investigated (Höglund et al., 1994). Chromosome 8. UPID 8 (pat) is apparently without any phenotypic effect, and one may suppose that this reflects a lack of imprinted genes on this chromosome. Benlian et al. (1996) had made the fortuitous discovery in an otherwise normal child with lipoprotein lipase deficiency, a recessive condition for which the locus maps to 8p22. Similarly, Karanjawala et al. (2000) discovered maternal isodisomy 8 by chance in a man participating in a diabetes research study. He was himself nondiabetic, although he did have the unusual history of a neuroendocrine gut tumor (carcinoid) at atypically young age. Chromosome 9. Maternal UPD 9 appears to be without effect (Björck et al., 1999; Engel and Antonarakis, 2002). Homozygosity at the SURF-1 locus due to isodisomy 9 is documented in twins with Leigh syndrome (Tiranti et al., 1999). Chromosome 10. Maternal UPD 10 appears to be without effect ( Jones et al., 1995). In uphd(10) mat with concomitant trisomy 10 mosaicism, it is presumably the trisomy rather than the UPD that causes a severe phenotype (Hahnemann et al., 2005). Chromosome 11. There are growth regulation loci in 11p15 that are expressed monoallelically, according to the parent of origin of the allele. These include the paternally expressed genes IGF2 and KCNQ1OT1, and the maternally expressed genes Uniparental Disomy and Disorders of Imprinting
•
359
H19 and CDKN1C. IGF2 and H19 are located within one of two “differentially methylated regions”4 (DMR1), such that IGF2 is only expressed from the paternal allele, and H19 only from the maternal allele. Similarly, KCNQ1OT1 (paternal expression) and CDKN1C (maternal expression) are under the control of the second region, DMR2 (Weksberg et al., 2010; Manipalviratn et al., 2009). Perturbation of these regions and genes can lead to two syndromes of opposite growth disorder: Beckwith-Wiedemann syndrome (BWS), of which overgrowth and hemihyperplasia are characteristic, and Silver-Russell syndrome (SRS), in which growth retardation and hemihypoplasia are key features. Beckwith-Wiedemann Syndrome. Mosaic segmental upd(11p)pat is the basis of about 20% of sporadically occurring BWS. In BWS, the striking clinical picture is that of overgrowth of tissues and organs. Thus, in upd(11p)pat, IGF2 and KCNQ1OT1 are expressed biallelically; and H19 and CDKN1C are silenced (“nulliallelic”). This imbalance is the basis of the excessive growth. Hemihyperplasia is a clinical indicator of this category, and those tissues with the greater fraction of upd(11p) cells may show a greater degree of overgrowth. Itoh et al. (2000) describe a child with BWS having a normal adrenal
(a)
(b)
(c)
gland on the right and a very enlarged one on the left: 30% of cells in the right gland had upd(11)pat, compared with 88% on the left. Epigenetic mechanisms exist due to other than UPD, noted in the later section on “Genetic Counseling” and as outlined in Figure 22–4 and Table 22–1. BWS due to 11p15 epimutation, affecting in particular the DMR2, has a particular association with IVF (Amor and Halliday, 2008; Lim et al., 2009; Manipalviratn et al., 2009). Paternal UPD 11 for larger extents of chromosome 11, and maximally the whole chromosome, may lead to typical or to more severe forms of BWS, or to a phenotype with severe intrauterine growth retardation, the differences likely reflecting tissue distribution of the UPD lineage (Grati et al., 2007). Silver-Russell Syndrome. SRS due to 11p anomaly can be considered the countertype to BWS, both clinically and at the molecular level (Schönherr et al., 2007). In SRS due to upd(11p)mat, or to 11p “epimutation” (hypomethylation of DMR1), the maternally active gene H19 functions biallelically, whereas IGF2 is underexpressed (Horike et al., 2009). Isolated hemihypoplasia, with shorter limbs on one side, has been recorded in association with epimutation (Zeschnigk et al., 2008). SRS can also be due to upd(7)mat, as noted earlier; the two
(d)
FIGURE 22–4 The no. 11 chromosomes in different chromosomal bases of Beckwith-Wiedemann syndrome (BWS). The maternal homolog is shown open, the paternal homolog is speckled, and the BWS critical region at 11p15 is shown cross-hatched. (a) The normal state of biparental inheritance of intact no. 15 chromosomes. (b) Paternal duplication of distal 11p. (c) Maternal reciprocal translocation disrupting the BWS critical region, with the other chromosome shown gray. (d) Mosaic segmental paternal uniparental disomy (UPD) of 11p, showing the chromosome 11 pairs of the two cell lines. The pair on the left shows paternal UPD for distal 11p (the speckled segments).
4 There is multiple nomenclature of these regions. DMR1 and DMR2 may be referred to as Imprinting Control Regions 1 and 2, ICR1 and ICR2. DMR1 is also known as H19 DMR, and the telomeric cluster; and DMR2 as KvDMR1, KCNQ1OT1 DMR, LIT1 DMR, and the centromeric cluster.
360
•
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
Table 22–1. Different Causes of Beckwith-Wiedemann and Silver-Russell Syndromes (see also Fig. 22–4) FRACTIONS (%) GENETIC FORM
BWS
Epigenetic Error on 11p15: Gain/loss of methylation 5 (gain) at DMR1 Loss of methylation at 50 DMR2 Uniparental disomy 20 (upd11pat) Chromosomal 11 Alterations: Duplication < 1 (pat) Paternal <<1 microduplication Maternal microduplication Maternal microdeletion <<1 Inversion, translocation < 1 CDKN1C mutation 10 Unknown 10
SRS
50 (loss)
5–10 (upd7mat) 1–2 (mat)
<<1
<<1 40
Note: Fractions (rounded) indicate relative frequencies; these data may be influenced by the clinical index of suspicion. DMR1 and 2, differentially methylated regions 1 and 2. DMR1 gain of methylation causes overexpression of IGF2 and nonexpression of H19. DMR2 loss of methylation causes overexpression of KCNQ1OT1 and nonexpression of CDKN1C (and see text). Sources: Algar et al., 2007; Manipalviratn et al., 2009; Eggermann et al., 2010a; Wakeling et al., 2010; Zollino et al., 2010a.
genetic forms have different underlying causes of the growth retardation (Binder et al., 2008). Wilms Tumor. In a study of 437 (nonsyndromic) Wilms tumor patients, Scott et al. (2008a) showed, in 13 of them, 11p15 abnormalities of the same sort that may be seen in BWS: upd(11p), and epimutations, a microinsertion, and a microdeletion in DMR1. A sibling of the child with a maternally inherited DMR1 microdeletion had a clinical diagnosis of BWS, illustrating that the identical genetic factor, although presumably with differing levels and
distribution of postzygotically arising mosaicism, can underlie the two disorders.5 Chromosome 13. Neither maternal nor paternal UPD 13, iso- or heterodisomy, appears to have any effect upon the phenotype (Berend et al., 1999; Soler et al., 2000). A unique example of familial UPD13, paternal and maternal, emphasizes this point: a normal mother with presumed 45,XX,i(13q)pat had a normal child with 45,XY,i(13q)mat (Slater et al., 1995). She may have been the result of monosomic rescue, and her son due to trisomic rescue! Chromosome 14. UPD 14 produces different syndromes according to the paternal or maternal basis of the disomy (Sutton and Shaffer, 2000; Engel and Antonarakis, 2002). Either may be seen in the setting of a normal karyotype or with a Robertsonian translocation (or “acrocentric isochromosome”). A balanced 45,der(13;14) Robertsonian translocation may reflect correction of an initially 46,der(13;14), +14 conception, while the 45,der(14;14) case might in fact result from a 45,–14 conception which then corrected by reduplication of the single chromosome 14 to give an i(14q) with isodisomy. Isodisomy may occur in the setting of a normal karyotype, and it may thus be less rare than is appreciated (Chu et al., 2004). Paternal UPD 14 is the more severe, with obstetric complication (polyhydramnios and premature labor), a particular pattern of malformation, growth retardation, and major functional neurological compromise (Sutton et al., 2003; Stevenson et al., 2004). Survival is poor. The bell-shaped thorax, reminiscent of Jeune syndrome, is a particular clinical pointer, and it has been observed at 23-week ultrasonography (Curtis et al., 2006); this anatomy may improve during childhood in those who survive (Chu et al., 2004; Kagami et al., 2005). Maternal UPD 14 produces a syndrome of pre- and postnatal growth retardation, a characteristic facies (to which the effects of an arrested hydrocephalus may contribute), and intellectual development may be “lownormal to normal.” Mitter et al. (2006) point out the overlap, to some extent, with the Prader-Willi neonatal phenotype. The critical segment, wherein the imprintable loci reside, is 14q32.2; the clinical picture is essentially similar in those with full or segmental UPD14 (Irving
5 In BWS with isolated DMR2 loss of methylation (the most common form; see Table 22–1), Wilms tumor is not seen, and this knowledge can inform clinical management.
Uniparental Disomy and Disorders of Imprinting
•
361
et al., 2010), and indeed the phenotype is also similar in cases with an epimutation (recognizable due to a marked hypomethylation at key loci within 14q32.2) (Zechner et al., 2009). Chromosome 15. Prader-Willi syndrome (PWS) and Angelman syndrome (AS) are the two UPD15 syndromes. It may be an oversimplification, but equally it may be a valid perspective, to think of these as each being caused by absent activity of a single genetic segment, or of a single gene, within 15q11– 13. In PWS, the basic defect may be absent activity of a transcript on the paternal chromosome 15, and which codes for a particular cluster (HBII-85) of small nucleolar RNAs (snoRNA) (Sahoo et al., 2008; de Smith et al., 2009). Different components of the PWS phenotype might therefore be mediated via perturbed functioning of different genetic targets of these snoRNAs. AS is due to absent activity of the UBE3A gene on the maternal chromosome 15. HBII-85 and UBE3A lie in close proximity on 15q11-q13, and both are under the influence of an imprinting control center (IC): from centromeric to telomeric on the chromosome, the order is IC, the HBII-85 cluster, and the UBE3A gene. The absence of activity is due either to the loss, or to the nonfunctioning, of this PWS/AS region on one chromosome 15 homolog. Loss is most commonly due to a simple interstitial deletion (“classical deletion”). Low-copy repeats on either side of the region can come together and set the stage for nonallelic homologous recombination (p. 296), leading to deletion of the PWS/AS region. Whether the phenotype comes to be PWS or AS depends upon which parent contributed the deleted chromosome. Nonfunctioning of (structurally normal) genes within 15q11q13 is due to the imprint status. This is most commonly the consequence of UPD 15, with the phenotype determined according to the parent of origin of the disomic pair of chromosomes. A rare cause is failure of, or damage to, the chromosome 15 IC. Study of these IC-damaged cases has cast much light on the processes of molecular pathogenesis in PWS and AS, and so the length of their commentaries that follow is quite out of proportion to their frequencies. In the case of AS, mutation in the UBE3A gene is a further category of mechanism. The 15q11q13 Imprinting Center. Normal persons have one paternally imprinted chromosome 15 and one maternally imprinted chromosome 15. The imprinting state of a chromosome 15 is set and reset
362
•
as it is transmitted down the generations, according to the sex of the transmitting parent. This resetting— an “epigenetic modification”—is dictated during gametogenesis from the cis-acting IC, with methylation of genes comprising, in large part at least, the crux of the process. The IC is bipartite, with a centromeric element, the AS-IC, and 35 kb distant a telomeric element, the PWS-IC, this latter including exon 1 of SNRPN. Interaction between these two elements directs the process. In maternal gametogenesis, the AS-IC has responsibility for initiating a paternal→maternal switch on the chromosome 15 that the mother herself had received from her father. The chromosome 15 she got from her mother retains a maternal imprint. With an active AS-IC, the UBE3A gene, lying about 1 Mb distant, is free to function, in the embryo to which this ovum gives rise. Vice versa, paternal gametogenesis serves to effect a maternal→paternal switch, or to retain a paternal status, on the chromosome 15 that the sperm contributes to the embryo. In consequence, a number of genes under its aegis are able to function, in part at least by being demethylated. The UBE3A gene’s activity is prevented. These epigenetic modifications operate only in cis, and so the maternal and paternal chromosomes continue to function autonomously, with their different repertoires of expression, during the life of the individual. A scheme for the various molecular defects of PWS and AS is presented in Figure 22–5; Horsthemke and Wagstaff (2008) provide a review. Table 22–2 sets out the test results for the different types of PWS and AS. Classical Deletion. This is the most frequent basis of the two syndromes, accounting for about 70% of both PWS and AS (Horsthemke and Buiting, 2006). The deletion removes 5.9 (class I) or 5.0 (class II) Mb within 15q11q13, encompassing the PWS and the AS genetic elements, and including the IC. There is one common distal breakpoint (BP3), and two variable proximal deletion breakpoint regions (BP1, BP2), due to duplicons at these sites. Nonallelic homologous recombination between the distal, and whichever proximal duplicon, then causes the deletions ( Ji et al., 2000). The behavioral phenotype is a little worse with the class I BP1-BP3 deletion than the class II BP2-BP3 deletion (Bittel et al., 2006). Larger deletions are infrequent and are associated with a more severe phenotype (Sahoo et al., 2007). If the deletion occurs on a paternally originating chromosome, it will cause the PWS phenotype to
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
FIGURE 22–5 An outline of the different genetic forms of Prader-Willi syndrome (PWS) and Angelman syndrome (AS). The PWS/AS critical region of chromosome 15 is depicted. A bipartite imprinting center with AS and PWS components (AIC and PIC) controls, in cis, the activity of a set of PWS genes and the UBE3A gene. A switched-on IC and an actively functioning gene are shown in unbroken line; a switched-off IC and an unactivated gene are shown in dashed outline. A mutated UBE3A gene is shown starred and with a dotted outline. (1) Normally, the UBE3A gene is transcribed only from the maternal chromosome (mat), and the PWS genes only from the paternal chromosome (pat), with each chromosome thus functioning appropriately for its parent of origin. In PWS there is nonfunctioning of the PWS genes because: (2) the PWS genes have been removed by a typical large deletion from the paternal chromosome; (3) both chromosomes are of maternal origin; (4) a microdeletion of, or mutation in, the PIC has fixed a maternal imprint status on the paternal chromosome. In AS there is nonfunctioning of the UBE3A gene because: (5) the UBE3A gene has been removed by a typical large deletion from the maternal chromosome; (6) both chromosomes are of paternal origin; (7) a microdeletion of, or mutation in, the AIC has fixed a paternal imprint status on the maternal chromosome; (8) there is a mutation in the UBE3A gene on the maternal chromosome. A further category (9) is not shown, comprising the 10%–15% in which no genetic defect can be shown. Approximate percentages of each PWS/AS category are indicated; in another ~10% of AS no genetic defect can be identified. patM, a maternally functioning chromosome of paternal origin; matP, a paternally functioning chromosome of maternal origin.
develop6; and vice versa, a maternal deletion produces AS. In a sense, there is an “unmasking of the silent elements” on the other chromosome. As well as the crucial PWS and AS genetic elements, a number of other loci may be deleted, and so the expression “contiguous gene syndrome” is not inappropriate, albeit having a somewhat different sense from its usage elsewhere in this book. One of the least important of these other loci is the P gene that contributes to normal pigmentation, and so children with PWS and AS due to classical deletion typically have fairer complexions than do their siblings.7 Mosaicism may lead to a milder phenotype (Golden et al., 1999; Tekin et al., 2000). In very rare cases of PWS with concomitant 47,XXY Klinefelter syndrome, the coincidence of the two conditions is likely to be fortuitous (Nowaczyk et al., 2004). Prader-Willi and Angelman Syndromes due to Deletion, Associated with Uncommon Rearrangement. Loss of the PW/AS region can be due to transmission of an unbalanced translocation or an inversion involving chromosome 15. The male carrier of a balanced reciprocal translocation in which one breakpoint is in the region of 15q13 can transmit an unbalanced complement to produce a deletion PWS child (Hultén et al., 1991; Smeets et al., 1992), and the female carrier can have a child with deletion AS (Stalker and Williams, 1998). There may be an additional effect from the concomitant imbalance involving the other chromosome of a translocation, such as the case in Torisu et al. (2004), a child who displayed features both of Angelman syndrome and the 1p36 deletion syndrome, due to a tertiary monosomy for these two segments, the mother being a balanced translocation carrier. A handful of PWS cases have been due to a Y;15 translocation with breakpoints in Yp and at 15q12-q13, deleting the PWS region, having the karyotype 45,X,der(Y),t(Y;15) (Vickers et al., 1994). A grandmother heterozygous for an inverted insertion of chromosome 15 had a PWS grandchild through her carrier son and an AS grandchild through her carrier daughter (Collinson et al., 2004). Uniparental Disomy and Prader-Willi Syndrome. About 30% of PWS is due to UPD (Horsthemke and Buiting, 2006). The cytogenetic study typically shows a normal 46,XX or 46,XY karyotype. Both
chromosomes 15 come from the mother, and so neither of the PWS critical regions is expressed. This functional lack causes the PWS phenotype. In most (80% or more), the UPD had its origin in a maternal meiosis I nondisjunction. A maternal age effect is clear: five times as many PWS children born to mothers under age 35 have a deletion as have UPD, but the reverse applies to those born to mothers 35 or over, in whom there is a five-fold excess of those showing UPD (Ginsburg et al., 2000). The phenotype is very similar to classical deletion PWS, although the facies may be less “typical” with the UPD form of PWS, and some of the minor manifestations are less likely to occur; in consequence, diagnosis may be delayed in comparison to deletion PWS (Cassidy et al., 1997; Gunay-Aygun et al., 1997). The UPD form of PWS is particularly associated with a psychiatric phenotype, typically presenting in young adulthood and characterized by a fluctuating psychosis and bipolar mood disorder (Verhoeven et al., 2003). A more severe form of UPD PWS is associated with a concomitant trisomy 15 mosaicism (Olander et al., 2000). Uniparental Disomy and Angelman Syndrome. Only about 1% of AS is due to UPD (Horsthemke and Buiting, 2006). As with PWS due to UPD, the karyotype is normal 46,XX or 46,XY. Both chromosomes 15 are from the father, and neither chromosome expresses the AS critical region. Most cases involve a postzygotic origin of the extra paternal chromosome, possibly following the “correction” of monosomy 15 due to a nullisomic ovum (as outlined earlier), and as with PWS, this is likely to reflect a maternal age effect. Very few AS children born to mothers under age 35 have UPD, but those born to mothers 35 or over have about equal numbers due to deletion and UPD (Ginsburg et al., 2000). A few are due to a paternal second meiotic error (Robinson et al., 2000). In parallel with the observations in UPD PWS noted earlier, the phenotype in AS due to UPD is not quite as severe as in the deletion form, with these children showing a lesser frequency of seizures, and some having a few words (Fridman et al., 2000). But it remains true that the handicap is severe. Prader-Willi and Angelman Syndromes Due to Uniparental Disomy, Associated with Chromosome 15 Rearrangement. Uniparental disomy can result from
6 An aide-mémoire: Prader-Willi due to Paternal deletion. 7 An additional copy of this gene leads to hyperpigmentation (Akahoshi et al., 2001). This is a nice example of a dosage effect: one copy of the P gene = pale skin, two copies = normal pigmentation, three copies = hyperpigmentation.
364
•
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
a variety of rearrangements involving chromosome 15. The male carrier of a reciprocal translocation involving chromosome 15 could transmit a disomic 15 spermatocyte from 3:1 nondisjunction, with the maternal chromosome 15 then being lost, and have a child with UPD AS; and vice versa, the female carrier could have a PWS child (Calounova et al., 2006; Heidemann et al., 2010). Similarly, a familial nonhomologous Robertsonian translocation in which one of the component chromosomes is a no. 15 giving a trisomic 15 conception, and with postzygotic loss of the chromosome 15 from the other parent, would lead to upd(15) with either PWS or AS, according to the sex of the carrier parent (Fig. 7–6) (Tsai et al., 2004). The same thing could happen if the translocation were de novo. A maternally originating de novo homologous der(15;15) (which may actually be a 15q isochromosome), with no chromosome 15 contributed from the father, would cause PWS (Robinson et al., 1994); and, vice versa, AS would result from a paternal isochromosome 15q (Tonk et al., 1996). Smith et al. (1994) describe AS from asymmetric segregation of a paternal 8;15 translocation (Fig. 8–5). The heterozygous father passed on his der(8) and his normal chromosome 15 (thus, paternal UPD), and there was absence of a maternal chromosome 15. Some PWS children with a 47,+idic(15) karyotype may actually have UPD of the two intact chromosomes 15, and the idic (15) is a phenotypically irrelevant relic of the original process of abnormal chromosomal behavior (Robinson et al., 1993). Imprinting Center Defects. A very small group of PWS and AS patients, about 1% and about 4%, respectively, have normal biparental inheritance and no classical deletion, but a uniparental pattern of methylation and gene expression (Horsthemke and Buiting, 2006). Most of these cases reflect abnormal function of the IC, while a minority, about 10%–15%, have an actual IC microdeletion. The latter category can be strongly suspected when there is a positive family history, while in the former, sporadic occurrence has been universally observed. Whether PWS or AS is seen depends upon which component of the IC is deleted or nonfunctional. Functional Imprinting Center Defect. Buiting et al. (2003) analyzed 44 PWS and 76 AS patients with a failure of IC functioning, an IC deletion or point
mutation having been excluded; these aberrant epigenetic states are referred to as epimutations.8 All cases were sporadic. Some shared with an unaffected sibling the 15q11-q13 haplotype on their paternal (PWS) or maternal (AS) chromosome, supporting the presumption of a de novo defect. With PWS, the basis of the epimutation may be a failure to erase the maternal imprint, as an act of omission (Buiting et al., 2003). Thus, for example, the father of such a PWS child passes on his maternal chromosome 15 with its maternal imprint still in place, and the child inherits two maternally imprinted no. 15 chromosomes. In AS, the typical scenario may be the imposition of an anomalous imprint status. This can be thought of as an act of commission: the mother inappropriately applies a paternal imprint to the chromosome 15 or fails to reset her paternal chromosome 15 that she passes to the child; or (since some maternal epimutations are mosaic) the error may occur postzygotically. If the error is incomplete, a milder AS phenotype may be seen (Brockmann et al., 2002). AS due to an imprinting defect, with loss of methylation of the maternal allele, may have an association with subfertility and artificial reproductive technology (Manipalviratn et al., 2009). If the association is indeed causal, the biological basis may be in the subfertility per se, or due to the superovulation treatment as part of IVF protocol, which leads to a failure to acquire normal UBE3A activation status in the ovum. Microdeletion of Imprinting Center. Microdeletions of the IC, generally of kilobase size, remove one or other of its major component parts, either the PWS-IC or the AS-IC. The inability to reset an appropriate imprint status leads to the “fixation of an ancestral epigenotype” (Saitoh et al., 1997). Only a handful of cases have been identified worldwide (Horsthemke and Buiting, 2006). Their particular importance to the counselor lies in the high recurrence risk: the mode of inheritance is essentially sex influenced—the parent’s sex, that is—autosomal dominant, with a 50% risk for the heterozygous father (for PWS) or the heterozygous mother (for AS), according to which component part of the IC is deleted. De novo mutations have been reported. In Prader-Willi syndrome due to IC microdeletion, the father would have received the deletion on his
8 The word mutation is normally taken to indicate that there is a change in the DNA sequence (from the Latin mutare, to change). By definition, no such change has occurred in an epimutation. But there has been a change in the functioning of the DNA.
Uniparental Disomy and Disorders of Imprinting
•
365
mother’s chromosome 15. He is normal, since an erased paternal imprint on his maternal chromosome is, naturally, correct. The deletion could have originated in his mother, or antecedent to her, provided transmission had been exclusively matrilineal. But when he passes this chromosome 15 with its fixed maternal epigenotype to a child of his, with the maternal→paternal imprint switch unable to function, the child has, effectively, a functional maternal UPD 15. Such a family is illustrated in Ming et al. (2000). Of 10 children, all of them normal and with normal karyotypes on standard cytogenetics, four inherited an IC microdeletion, presumably from their deceased mother (their father was proven not to have the deletion). Two of these children were male, and each went on to have, in the next generation, a child with PWS: an example of “grandmatrilineal inheritance.” The laboratory demonstration of an IC deletion is complex, in concept and in practice (Buiting et al., 2003). In Angelman syndrome due to IC microdeletion, the scenario is essentially the obverse of the above. A microdeletion on the maternal chromosome 15 removes the AS-IC. The defect may have arisen de novo from the maternal grandfather of the AS child, or alternatively, there could have been patrilineal transmission of the mutation, harmlessly, for any
number of previous generations. Transmission from the grandfather to the mother would be without phenotypic consequence, since a paternally originating chromosome 15 would in any event have its AS-IC inactivated. But in oögenesis in the mother, the normal paternal→maternal switch on the abnormal chromosome cannot be effected (thus, “fixation” of the ancestral paternal epigenotype). If the child receives this chromosome 15 from the mother, both homologs carry a paternal imprint. In consequence, the child has AS. Two such Japanese families, independently ascertained and reported, had exactly the same 1.487 Mb deletion, from nucleotides 22,938, 518 to 24,425,426, and may well have represented distant branches from the same, presumably male, ancestor (Sato et al., 2007). Angelman Syndrome due to UBE3A Gene Mutation. Classical point mutation, affecting the UBE3A (ubiquitin protein ligase 3A) gene, is an important contributor to AS etiology (Abaied et al., 2010). This gene is expressed from both parental chromosomes in some tissues, but, in the brain, from only the maternal chromosome. The (normal) paternal allele does not function in embryonic brain, or at least in particular parts of the brain. Thus, if the maternal gene is mutated, there is no UBE3A expression, and in consequence brain development is
FIGURE 22–6 A family with inherited Angelman syndrome, due to a UBE3A mutation, reported in Moncla et al. (1999). Filled symbol, Angelman syndrome; bull’s eye symbol, mutation carrier, demonstrated or inferred; N, demonstrated noncarrier. Note that all the affected children are born to carrier mothers, but that these mothers are related to each other through the male line. Some normal children have been proven to be noncarriers with molecular testing (N in symbol), but the reader can also determine that any unaffected child of a potential carrier mother, such as IV:1 and 2, the children of III:4, or V:9, the sibling of an affected child, cannot be carriers. An inherited imprinting center mutation could present a similar pedigree. 366
•
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
compromised (Rougeulle and Lalande, 1998). In a mouse knockout model, Ube3a expression was compromised in certain cells of the hippocampus, a crucial structure in learning and memory, and of the cerebellum, which may have a role in learning as well as its classic role in coordination (Albrecht et al., 1997). The human situation is quite likely to be similar. (Mouse knockout models for PWS are lethal.) About 70% of inherited “nondeletion non-UPD non-IC” AS is due to UBE3A mutation of maternal origin. As for sporadic AS, and considering those patients with normal methylation results, a UBE3A mutation is seen in about 30%. The severity of phenotype in the mutation form falls between the deletion and UPD cases (Abaied et al., 2010). Multigenerational transmission may be seen, with the revealing observation that AS children are born only to carrier daughters of carrier males (Fig. 22–6). The mutation transmitted by the father has no effect in his child, since this chromosome 15 region would in any event carry a paternal imprint and be silenced. Angelman Syndrome with No Deletion, No Uniparental Disomy, No Imprinting Mutation, and No UBE3A Mutation In some 15%–20% of AS, no genetic defect can be found ((Hitchins et al., 2004; Horsthemke and Buiting, 2006). There is a normal karyotype, with no deletion demonstrable on fluorescence in situ hybridization (FISH), normal methylation analysis (at least on the sampled tissues), biparental inheritance, and an apparently intact UBE3A gene. There may be an epigenetic influence whereby a normal UBE3A gene on the maternal chromosome fails to activate normally during embryogenesis. Or there may be some other AS genetic basis, as yet unknown. Chromosome 16. This is one of the more commonly seen UPDs, and it is almost always due to correction of trisomy 16 of maternal meiotic origin. Thus, it is typically a maternal UPHD. It has been difficult to separate out the effects of the UPD, and of a placental insufficiency due to confined placental mosaicism for trisomy 16, this typically being the route by which the UPD comes to be recognized, following chorionic villus sampling; and in addition, a possible residual occult fetal trisomy mosaicism always remains as a potential confounder. Yong et al. (2002) showed in a large series of mosaic trisomy 16 discovered at prenatal diagnosis, that the degree of fetal growth restriction, and probably the malformation rate, was greater in those with upd(16)mat than in those with biparental inheritance, thus suggesting a
role of the UPD per se. A single case is recorded of body stalk defect in a fetus with upd(16)mat (Chan et al., 2000). As for paternal UPD 16, it seems probable that it has no clinical consequences (Engel and Antonarakis, 2002). The usual rare risk from isozygosity for a recessive gene applies, as exemplified in Wattanasirichaigoon et al. (2008), who report a child with hydrops fetalis due to hemoglobin Bart’s, consequential upon upd(16)mat. Chromosome 17. Two cases of complete upd(17) mat have been described. One 46,XY child was normal, ascertainment having been via the discovery of trisomy 17 mosaicism at amniocentesis (Genuardi et al., 1999). Lebre et al. (2009) identified the UPD in an infant with cystinosis, a recessively inherited multiorgan storage disease, the locus of which is on chromosome 17p, and this segment being in isodisomic state in the child. The abnormal phenotype in a child with a segmental upd(17)mat, involving 17q25.3, may be due to some other factor (Rio et al., 2001). Chromosome 20. A prima facie case exists for a UPD 20 syndrome. Eggermann et al. (2001) review three reported cases of upd(20), one paternal and two maternal, with a major malformation phenotype in the former, and growth retardation the common observation in the latter two. A causal link is possible, as is also true concerning two abnormal children initially ascertained because of trisomy 20 mosaicism at amniocentesis (Salafsky et al., 2001; Velissariou et al., 2002). A single sporadic case of pseudohypoparathyroidism in association with a paternal upd(20)pat (UPD for the long arm) might reflect a true effect of the UPD, since the causative gene (GNAS1) on 20q is known to display a parent-oforigin effect (Bastepe, 2008). This abnormality probably arose as a mitotic event, according to the scenario as set out in Figure 22–3d. Further support for this view comes from Aldred et al. (2002), who studied two children with deletions of 20q, one of maternal and the other of paternal origin, in whom the different patterns of parathyroid endocrinology were consistent with what is understood of the complex behavior of the GNAS1 gene. Chromosome 21. UPD 21, maternal or paternal, appears to be without effect (Engel and Antonarakis, 2002). Chromosome 22. Maternal UPD 22 has generally not been causally associated with any defect Uniparental Disomy and Disorders of Imprinting
•
367
(Kotzot, 1999; Engel and Antonarakis, 2002). Intrauterine growth retardation, if present, may more likely reflect the influence of a trisomic 22 placenta, or low-level “occult” mosaicism of the fetus (Balmer et al., 1999; Bryan et al., 2002). A child with UPD 22 due to “balancing” recombinant chromosomes 22 from parental inversions was developing normally at age 20 months (Kariminejad et al., 2011, and see p. 171).
maternal set (triploidy, with a total chromosome count of 69), a partial hydatidiform mole results. Hydatidiform mole is discussed in more detail in Chapter 23. UPDmat of an oöcyte, following failure of a premeiotic or of a meiotic cell division, leads to benign cystic ovarian teratoma, an unusual tumor of the ovary in which several embryonic tissues may be represented (Miura et al., 1999).
Chromosome X. Neither upd(X)mat nor upd(X)pat appears to have any consequence in the 46,XX person, with the usual exception of homozygosity for a recessive mutation (Quan et al., 1997). However, there may be a subtly different neuropsychological phenotype according to the parent of origin in monosomy X (self-evidently a uniparental condition). In a British study, 80 girls with Turner syndrome underwent behavioral evaluation, 55 of whom were 45,XM and 25 were 45,XP. The 45,XP girls were more socially adept and more articulate than the 45,XM girls. Speculatively, this may represent the effect of an imprintable X-borne “locus for social cognition” that is functional on the X chromosome transmitted from a father, and nonfunctional on the X from a mother (Skuse et al., 1997). Autism, which is a male-susceptible condition, is associated with 45,XM in the case of autistic girls with Turner syndrome (Donnelly et al., 2000). In terms of response to growth hormone, it makes no difference whether the child is 45,XM or 45,XP (Tsezou et al., 1999). Upd(X)pat offers the intriguing scenario of father-to-son transmission of an X-linked gene. A 24,XY gamete from a hemizygous father could produce a 47,XXY zygote, which could subsequently lose the maternally contributed X; or the ovum could be nullisomic X, with sex chromosomal complementation producing 46,XY. Either mechanism could explain the observations in a family with the X-linked form of ectodermal dysplasia, as presented in Ferrier et al. (2009).
Mosaicism for Complete Uniparental Disomy. Abnormal cytogenetic events around the time of fertilization—such as two sperm entering an ovum to produce a zygote with three pronuclei, or an ovum undergoing a mitosis, and then cell lines of different (but diploid) genetic constitution being produced—can be the basis of a mosaicism for UPD. This can be UPDmat/biparental or UPDpat/ biparental mosaicism, and it may be confined to the placenta or involve the fetus as well. Complete UPDpat/normal mosaicism in the placenta (androgenetic/biparental mosaicism) leads to the histological phenotype of mesenchymal dysplasia (discussed in more detail on p. 396). If the UPDpat line also involves the fetus, features of Beckwith-Wiedemann syndrome may be present, and according to the cellular distribution of the UPD tissue, a more complex clinical picture may be observed (Wilson et al., 2008; Jalil et al., 2009). Two unique cases cast light on how aberrant chromosomal behavior in the perizygotic period can lead to UPD-related pathology. A 46,XX/46,XY male child described in Strain et al. (1995) with growth asymmetry had complete maternal isodisomy in the 46,XX cell line and biparental inheritance in the 46,XY line. It may be that an ovum had completed a mitosis on its own, and then one of its daughter cells received the sperm (for the 46,XY line) while the other underwent endoreduplication (for the 46,XX,upd(mat) line): thus, biparental/gynogenetic mosaicism. A diploid sperm may have been the basis of the case in Hsu et al. (2008), in which the pregnancy from an apparently normal IVF embryo ended in intrauterine fetal death, with a portion of the placenta of molar appearance, at 14 weeks. Three separate genetic constitutions could be determined. The molar tissue was androgenetic 46,XX,uphd(pat); the fetus and placenta, both 46,XX, shared the same maternal genome but had different paternal genomes. Any explanation for this circumstance is necessarily complex.
Uniparental Disomy for the Entire Chromosomal Complement (Complete Uniparental Disomy) Nonmosaic paternal uniparental disomy (UPDpat) for the full diploid complement—all 46 chromosomes are of paternal origin—produces the placental disorder of complete hydatidiform mole. When, in addition to a double set of paternally derived chromosomes, there is also a haploid 368
•
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
GENETIC COUNSELING Uniparental Disomy for Individual Chromosomes No instance of recurrence of full UPD for a particular chromosome, with a 46,XX or 46,XY karyotype, is known, and we assume there to be no discernibly increased recurrence risk. The association with increasing childbearing age is to be noted, but in reality the increase in risk for older mothers would be very small.
Segmental Uniparental Disomy Segmental UPD arising postzygotically, and which is karyotypically 46,XX or 46,XY, we presume to imply no increased risk. UPD due to rearrangement would have a risk according to the nature of the specific rearrangement.
Four “Imprinting Syndromes” with More Than One Genetic Basis BECKW ITH -W IED EMA NN SY NDROM E
The considerable majority (about 85%) of BeckwithWiedemann syndrome (BWS) occurs sporadically, including the two more common categories of UPD 11 and epigenetic error. The other categories that may have an important recurrence risk are recognized either by an abnormal cytogenetic report and/or by a positive family history. A detailed treatment is given in Weksberg et al. (2010). Uniparental Disomy 11. About a fifth of sporadic cases are due to mosaic segmental paternal UPD of 11p. This category of BWS can be suspected clinically if there is hemihyperplasia. No increased recurrence risk applies in the setting of segmental UPD and a normal karyotype. Epigenetic Error. In sporadic BWS with biparental disomy, the underlying cause is an epigenetic error (“epimutation”) affecting the ovum or early conceptus. This is the basis of a little over half of all cases. There is biparental inheritance with aberrant methylation on the maternal chromosome of either DMR1 (gain of methylation, ~5% of cases) or DMR2 (loss of methylation, ~50% of cases), the latter combination particularly associated with IVF. No cases of recurrence in this setting are known, and this fits the understanding of a typical postzygotic
generation of the epimutation (Scott et al., 2008b) (but note recurrences in the section on “Epigenetic Silver-Russell Syndrome”). Theoretically, there might be a very small increased risk, if the same susceptibility factors (subfertility, IVF) were operating. 11p Rearrangement. Chromosome rearrangements are rare causes of BWS. A balanced reciprocal translocation or an inversion with one breakpoint in distal 11p, if of maternal transmission, may lead to BWS (Li et al., 1998). An unbalanced distal 11p15 duplication, if of paternal origin, leads to double expression of IGF2 in the 11p15 region, and this brings about the growth pattern of BWS (and if of maternal origin, a Silver-Russell growth retardation phenotype results; see later). Functional trisomy of nonimprinted 11p segments, or other imbalance due to a translocation, may contribute to the clinical picture (Han et al., 2006; Russo et al., 2006; South et al., 2008c; Bliek et al., 2009). The recurrence risks for these various circumstances will depend upon the nature of the rearrangement, and the parental karyotypes. Mendelian Mutation. Autosomal dominant BWS accounts for about 10% of cases, the major locus CDKN1C. Typically, only the offspring of female heterozygotes are affected. Careful review of the pedigree in the maternal line is necessary to identify mildly affected individuals, and bearing in mind the amelioration of phenotype with time (Hunter and Allanson, 1994; Elliott et al., 1994). One might consider the very rare case of deletion of the differentially methylated region DMR1 also to be in the category of Mendelian mutation; a child receiving this deletion from a mother has BWS, due to consequential biallelic IGF2 expression (Sparago et al., 2004). A rare recessive basis of a maternal susceptibility to have a BWS child resides in the NLRP2 gene, with failure to impose a proper imprint upon ova (Meyer et al., 2009). SI LV ER- R USSEL L SYN D R O M E
Most SRS can be traced to an anomaly of chromosome 7 (UPD) or chromosome 11p15 (epimutation), the latter somewhat mirroring the mechanism in BWS, as outlined earlier. Both these genetic forms typically imply a low risk of recurrence (Eggermann et al., 2010a). Chromosome 7. Upd(7)mat is seen in up to 10% of cases, and the clinical phenotype in this group Uniparental Disomy and Disorders of Imprinting
•
369
is typically more severe (Binder et al., 2008). Sporadic occurrence has been the universal observation. Chromosome 11. Epimutations of 11p15, with hypomethylation of the IGF2/H19 differentially methylated region (DMR1), comprise the largest single category: according to the stringency of clinical criteria, these account for about half of all SRS. This category may be associated with conception by IVF (Wakeling et al., 2010). Sporadic occurrence is very much the rule, albeit that very rare recurrences are on record, due to parental heterozygosity or parental gonadal mosaicism (Bartholdi et al., 2009). Structural rearrangement of 11p15, such as microduplication involving the differentially methylated region DMR2, is a rare cause (Eggermann et al., 2009). A familial translocation, such as the t(11;15) (p15.5;p12) described in Eggermann et al. (2010c), in which one segment comprises distal 11p, could lead to either SRS, if maternally transmitted, or BWS, if from the father (this is analogous to the
scenario noted later for 15q11q13 rearrangements, with different phenotypes according to the parent of transmission). SRS Phenocopies. A SRS-like clinical picture can result from genomic changes elsewhere on the karyotype (Bruce et al., 2010). PRA D ER-W I L L I SYN D R O M E
A summary of the different genetic forms of PWS, and the associated risks of recurrence, is set out in Tables 22–2 and 22–3. Classical Deletion 15q11q13. The empiric observation of zero recurrences out of some thousands of “trials” underscores the considerable unlikelihood of significant paternal gonadal mosaicism for the deletion observed in the PWS child. This is the basis of the substantial optimism that can be offered to parents in terms of any further pregnancies.
Table 22–2. Assessment of Genetic Category of Prader-Willi and Angelman Syndromes According to Results of Cytogenetic and Molecular Testing METHYLATION PATTERN OF NO. 15s
PARENTAL ORIGINS OF NO. 15s
+
mat
bi
N ±
mat mat
mat bi
N
mat
bi
+
pat
bi
deleted
N N
pat pat
pat bi
“intact” “intact”
N
bi
bi
mutated
N
bi
bi
“intact”
CYTOGENETICS FISH
PraderClassical ± Willi deletion syndrome upd(15)mat N Imprinting N center microdeletion Other imprinting N center defect Angelman Classical ± syndrome deletion upd(15)pat N Imprinting N defect UBE3A N mutation Epigenetic error N
UBE3A GENE
Notes: A normal cytogenetic/FISH result is indicated by N, and an abnormal result by +. Most classical deletions are detectable cytogenetically, but a few may be missed (indicated as ±). Methylation patterns/parental origins: FISH, fluorescence in situ hybridization; mat, maternal; pat, paternal; bi, biparental. “Intact” means that the DNA sequence of the gene is normal, but its function is epigenetically compromised.
370
•
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
Table 22–3. Approximate Relative Frequencies and Recurrence Risks, to Parents Having Had an Affected Child, for the Different Categories of Prader-Willi and Angelman Syndromes CATEGORY
RELATIVE FREQUENCY
RECURRENCE RISK
PraderClassical deletion Willi upd(15)mat syndrome Imprinting center defect 15q translocation/inversion
70% 25% 2%–5% Rare
Extremely low* Extremely low* 50% According to rearrangement
Angelman Classical deletion syndrome upd(15)pat Imprinting center defect UBE3A mutation 15q translocation/inversion Epigenetic error/unknown
70% 3%–5% 2%–5% 10%–15% Rare 10%
Extremely low** Extremely low* 50% 50% According to rearrangement Presumed very low
*No case yet recorded. **Only one case in the world recorded (Kokkonen and Leisti, 2000).
A figure of around 0.1% may be a fair one to offer for the risk of recurrence. Nevertheless, the theoretical possibility of paternal gonadal mosaicism, or of a paternal predisposition to undergo chromosome 15 deletion in spermatogenesis (Molina et al., 2010), obliges acknowledgment that the risk is not zero. If prenatal diagnosis is pursued, CVS can be offered using FISH for deletions, or the SNRPN methylation test (Buiting et al., 1998). Uniparental Disomy 15, Karyotype 46,XX or 46,XY. We know of no recorded instance of recurrence of upd(15)mat PWS in a chromosomally normal couple, and we would otherwise assume, on theoretical grounds, any increased risk in a future pregnancy to be practically negligible, the modest maternal age effect notwithstanding. Functional Defect (“Epimutation”) of PraderWilli Syndrome Imprinting Center. These extremely rare cases of IC defects will require individual expert advice. They can be suspected if a child has typical PWS clinically, but there is neither classical deletion nor UPD demonstrable. All cases of functional IC deficiency have so far been sporadic (but very few are known). Prader-Willi Syndrome Imprinting Center Microdeletion. The recognition of these cases will require referral to a specialist laboratory. A positive
family history, if observed, would oblige the assumption of this category, unless or until otherwise proven. Assuming the father carries the genetic defect, the recurrence risk is high, namely 50%. SNRPN methylation testing on CVS can identify an affected pregnancy. The father’s brothers would have a 50% likelihood to be heterozygous (making the assumption that their mother would carry the mutation), and in that case, these brothers would also have a 50% risk to have a PWS child. Equally, his sisters could be carriers, but their children would all be unaffected, and it would only be their sons who might, in the next generation, have the risk for a PWS child. The siblings of the affected child would themselves have no different genetic risk than the general population. The reader should work through the reasoning behind these various risk assessments, even though most counselors will never encounter this actual circumstance in the clinic. Uncommon Cytogenetically Detectable Rearrangement. The nature of the rearrangement (see the section on “Biology”), and the parental karyotypes, will determine the recurrence risk in each type. PWS Phenocopies. A duplication of the segment Xq21.1–q21.31 may lead to a clinical picture very reminiscent of PWS in the older child, while deletions distal to Xq25 are associated with a phenotype Uniparental Disomy and Disorders of Imprinting
•
371
more resembling infantile PWS (Gabbett et al., 2008). A small fraction of males with fragile X syndrome present a PWS-like phenotype (p. 246). A focused Xq analysis, and a fragile X study, may thus be warranted in patients in whom a diagnosis of PWS is suspected on clinical grounds, but in whom chromosome 15 tests are normal. Deletion at 6q16.2, and the 1p36 deletion, may also be associated with a PWS-like clinical picture (D’Angelo et al., 2006; Varela et al., 2006; Bonaglia et al., 2008). ANG E LMA N SY N D R O M E
A summary of the different genetic forms of AS, and the associated risks of recurrence, is set out in Table 22–2. More detail is available in the reviews of Stalker and Williams (1998), Clayton-Smith and Laan (2003), and Van Buggenhout and Fryns (2009). The clinical diagnosis of AS is sometimes easy (parents have recognized the condition in their child having seen a television program), but at other times, more challenging. Of course if accurate genetic advice is to be given, an accurate clinical diagnosis is crucial. The possibility of Rett syndrome may need to be considered (Scheffer et al., 1990). A proposed very simple clinical screening test is to gauge the response to a vibrating tuning fork held close to one ear: an AS child will laugh and turn toward it (Hall and Cadle, 2002). The counselor must take the trouble to obtain a detailed family history. A genetic defect could have been transmitted through males for some generations, and only causing AS when it had been passed from a daughter of such a male. Figure 22–6 shows a family in which some quite distant relatives, including second cousins once removed and first cousins twice removed, had AS due to an inherited UBE3A mutation. Classical Deletion 15q11q13. Similarly to PWS, about 70% of AS is due to a de novo interstitial deletion. Only one case in the world is recorded of recurrence in siblings of a typically sized deletion. This case involved, presumably, a mother with gonadal mosaicism (Kokkonen and Leisti, 2000). Thus, as for classical deletion-PWS, we presume a very low— but clearly not zero—recurrence risk. There are two recorded examples of deletion AS in cousins, which manifestly represented coincidental de novo events in these families, in that different ancestral chromosomes were involved (Connerton-Moyer et al., 372
•
1997). The comments on prenatal diagnosis in PWS (see earlier) apply similarly here. Uniparental Disomy 15, Karyotype 46,XX or 46,XY, Parents’ Chromosomes Normal. AS due to paternal UPD 15 is rare; as discussed earlier, the initial error may actually reflect a maternal age effect. Interestingly, the AS phenotype may be somewhat milder in UPD15, and in some children it was only after an electroencephalogram (EEG) showed typical findings that the diagnosis was suspected (Bottani et al., 1994). But this does not mean that some upd(15)pat AS children may not be severely affected (Prasad and Wagstaff, 1997). No recurrence is on record (Chan et al., 1993), and we assume on theoretical grounds that no usefully measurable increased risk would exist. Angelman Syndrome Imprinting Center Microdeletion. Assuming the mother carries the genetic defect, there is a high recurrence risk, namely, 50%. SNRPN methylation testing on CVS can identify an affected pregnancy. The possibility of maternal gonadal mosaicism for an IC mutation complicates the picture (Stalker et al., 1998). As noted earlier, but vice versa for AS, the siblings of the carrier mother could also be carriers (assuming their father to be heterozygous). However, it would only be the sisters who would have the risk for an AS child. Functional Defect (“Epimutation”) of Angelman Syndrome Imprinting Center. The previous comments on PWS apply similarly here. All cases of AS due to a functional IC defect have so far been sporadic, but it would be prudent to offer prenatal diagnosis in a subsequent pregnancy (SNRPN methylation testing). Although the numbers are very small (but epimutation-AS is rare), there are grounds for supposing there might be a link with infertility/IVF (Manipalviratn et al., 2009). Had there been such a reproductive history, this fact would need to be weighed. UBE3A Mutation. If the mother carries the mutation, the risk for recurrence is 50%. Maternal mosaicism has been recognized (Malzac et al., 1998; Hosoki et al., 2005), and so nondemonstration of the mutation in the mother does not necessarily exclude a genetic risk. Indeed, it may be that such mosaicism is not uncommon (Stalker et al., 1998). It may be appropriate to track the mutation through the patrilineal family, in order to be able to offer
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
genetic counseling to female cousins who might be carriers. The reader should study the illustrative pedigree in Figure 22–6. There is the practical point that routine clinical testing for UBE3A is available in only a very few laboratories, and at some cost. No Genetic Defect Demonstrable. In a small fraction of AS, about 10%, no cytogenetic or molecular defect, nor UBE3A mutation, is demonstrable. Some of these cases could conceivably reflect a mutation that has not been able to be detected. The family history, if positive, may compel the assumption of a mutation, and thus imply a high recurrence risk. A negative family history might support the inference of a low risk, but it would not allow a definite assumption. If a normal sibling carried the same 15q11q13 haplotype, using DNA markers, a low-risk scenario would be probable. Expert advice should be sought. Uncommon Cytogenetically Detectable Rearrangement. The nature of the rearrangement (see the “Biology” section), and the parental karyotypes, will determine the recurrence risk in each type. The rare circumstance of UPD associated with a parental Robertsonian translocation is noted on p. 152. Angelman Syndrome Phenocopies. Rett syndrome has considerable clinical overlap with AS, as do the 22q13.3 deletion and Pitt-Hopkins syndrome (Hitchins et al., 2004; Takano et al., 2010). A S I M P LIF IC ATIO N F OR ANG E LMA N SY N D R O M E
Some parents will not find it easy to come to grips with these various possible causes for their child’s condition even if, in the end, they need only consider the category that applies to themselves. It may be helpful to discuss AS, and the risks of recurrence, in the following terms. Let us say that AS is due, simply, to a lack of the UBE3A protein, a very important protein that is necessary for the brain to grow normally. The gene for UBE3A works only on the chromosome 15 from the mother, while the gene on the father’s chromosome is dormant. There is a switch on the mother’s chromosome that makes this gene work. • If the bit of the maternal chromosome that contains this gene is missing (deletion), or if the mother’s chromosome is replaced by another one
from the father (UPD), no UBE3A protein can be made. These two types happen as one-off events. • If the switch fails on the mother’s chromosome, then the gene remains dormant, and no protein is made (imprinting center fault). This type can happen one-off, as though the switch “gets stuck,” for reasons that we do not well understand. Or there may be a genetic fault in the actual switch, and in this case the defect could be passed to a subsequent child. • If the UBE3A gene itself is faulty on the mother’s chromosome (mutation), no protein is made, or only an abnormal protein that cannot function. The genetic risk depends on whether the faulty gene started with the child (no increased risk) or if the mother is a carrier (high risk). Note that the mother can be a carrier and still be perfectly normal, since the faulty gene would be the one she got from her father, and so in any event it would be switched off. • Sometimes the UBE3A gene fails to work, even though the maternal chromosome is normal and has a normal switch. We do not know why this happens (there has been a suggestion that one cause may be if there had been difficulty achieving the pregnancy, either naturally or with IVF). This type is a one-off event. A common question parents have is whether their normal children might, in the next generation, have an AS child. Or the aunts and uncles of an AS child might want advice about risks to their future children or to their grandchildren. The answers are as follows. • The normal siblings of an AS child have no increased risk, for any genetic category, with the possible exception of a familial translocation. Even if the AS child has (or had) a potentially heritable type of UBE3A genetic defect, the fact that the sibs themselves are normal declares that they cannot have received it. If they had got the abnormal gene, they would have AS; since they do not have AS, they cannot have the gene. The sex of the siblings is immaterial. • Aunts and uncles have an increased risk for children or grandchildren of theirs only if a heritable type of AS is involved (imprinting center defect, UBE3A mutation). In that case, an uncle could be a carrier, but his children would not be at risk, since the UBE3A gene would be dormant anyway. Daughters of his, however, Uniparental Disomy and Disorders of Imprinting
•
373
could have an AS child. A carrier aunt would have a high risk (50%) to have an AS child. But her grandchildren, through her normal sons and daughters, would have no increased risk. Her normal children would have declared themselves, by their very normality, not to have inherited the genetic defect. P RE NATAL TESTIN G CONSI DE RAT I ONS
The American College of Medical Genetics has recommended testing for UPD be offered when an imprintable chromosome (6, 7, 11, 14, 15) has raised a concern at prenatal diagnosis, either with level II mosaicism, or especially in the context of a Robertsonian translocation, or if there are indicative
374
•
ultrasound findings (Shaffer et al., 2001). We note that this advice is somewhat more liberal than that of Kotzot (2008b), who has suggested that only chromosome 14 and 15 be considered in this regard (and see p. 456). U N I PA R EN TA L D I S O MY F O R T H E EN T I R E C HR O M OS O M A L CO M PL EM EN T
UPD for the entire paternal chromosome set (hydatidiform mole) is associated with an increased recurrence risk; this is discussed in detail in Chapter 23 (p. 400). There is no discernibly increased risk for the recurrence of UPD for the entire maternal chromosome set (ovarian teratoma).
D I S O R D E R S A S S O C I AT E D W I T H A B E R R A N T G E N O M I C P R I N T I N G
PART SIX REPRODUCTIVE FAILURE
This page intentionally left blank
23 gametogenesis and conception, pregnancy loss and infertility
HUMAN CONCEPTION and pregnancy is both a vulnerable and a robust process. Vulnerable, in that a large proportion of all conceptions are chromosomally abnormal, with the great majority of such pregnancies aborting. Robust, in that more than 99% of the time, a term pregnancy results in a chromosomally normal baby. Unbalanced chromosomal abnormalities are seen in less than 1% of newborns (see Table 1–3 in Chapter 1). But the economic cost of chromosomally abnormal conceptions is not horrendous; it is measured largely in terms of miscarriage, seen or unseen. The occasional chromosomally abnormal child is, relatively speaking, an exceptional outcome—the tip of an iceberg (Fig. 23–1). Most of this chromosomal vulnerability lies in the process of producing eggs and sperm. Meiosis hangs, literally and figuratively, upon “tender filaments,” and
often the meiotic chromosomes are incorrectly distributed to the daughter cells. Indeed, humans are more prone to produce aneuploid germ cells than any other species studied (McFadden and Friedman, 1997).1 The group who are particularly likely to produce abnormal gametes are carriers of balanced chromosome rearrangements, and much of this book is devoted to that fact. Advances in reproductive technology now enable many otherwise infertile couples to have children. Translocation carriers may have recourse to preimplantation genetic diagnosis (PGD) as a means to improve their chances of achieving a successful pregnancy (Chapter 26). In the case of men with poor sperm production, intracytoplasmic sperm injection (ICSI) at in vitro fertilization (IVF) is a means to get a single sperm into an egg. Success with
1 Warburton suggests that this error rate conveyed an evolutionary advantage in former times: miscarriage due to aneuploidy led to a wider spacing of offspring, allowing a woman to devote scarce resources to a more manageable number of offspring, more of whom would survive to contribute their genes to the following generation. What was beneficial in a prehistoric society has been quite the opposite for many women, now hostage to their biology, in the present century (a concept not without several other examples).
•
377
FIGURE 23–1 The iceberg of chromosomal pregnancy loss.
IVF is not necessarily easy to achieve, neither is it a certain outcome, and counselors dealing with infertile couples need a particular awareness of the psychological and practical difficulties they may face (Boivin et al., 2001). A “failed embryo transfer” following IVF may be considered as a form of pregnancy loss not unlike that of the natural miscarriage of a wanted pregnancy.
BIOLOGY Gametic Cytogenetics Many more sperm are made than eggs, by orders of magnitude, and logically one might have expected a higher standard of meiotic fidelity in the scarcer gamete (Hunt and Hassold, 2002). But in fact it is the other way round, and so it is the egg that commands most of our attention in terms of the practical relevance of gametic chromosomal pathology. OÖCY TES AN D P O LAR BODI ES
IVF is widely applied in the management of infertility, and one consequential benefit of this has been the access afforded to study of the oöcyte and its minor partner, the polar body. Many eggs sampled prove to be surplus to the requirements of the couple, and they are often willingly donated for research. 378
•
R E P R O D U C T I V E FA I LU R E
Pellestor et al. (2006) have reviewed the literature, in which different groups applied various methodologies; and Fragouli et al. (2006b) report a fourcity (Dundee, London, Rochester, and New Haven) collaboration, in which comparative genomic hybridization (CGH) was applied, with the view that this technique would be less prone to artifactual misinterpretation. Overall, a fifth to a quarter of oöcytes are cytogenetically abnormal. In the CGH study, many eggs and polar bodies could be analyzed as a pair, allowing the detection of reciprocal imbalances to function as an internal control. Most of the abnormalities are accounted for by hyperhaploidy with a 24-chromosome count (an additional double-chromatid or single-chromatid chromosome), hypohaploidy with a 22-chromosome count (a missing homolog) or a 23-chromosome count that could be described as “22½” (one homolog represented by only a single-chromatid chromosome), and diploidy with 46 chromosomes. In order to address what might be the basis for these abnormalities, Cupisti et al. (2003) analyzed a set of eight chromosomes (large, medium, and small representatives), from pairs of oöcytes and their first polar bodies. (These were spare eggs, 236 in all, from women presenting for infertility treatment due to a variety of causes, both male- and female-based.) The large chromosome (1 being the chosen representative) had not misdivided, and only the X of the three medium-sized chromosomes (9, 12, X) had not. Of the smaller chromosomes (13, 16, 18, 21), all were seen in aneuploid state, and for each chromosome, 2%–3% of eggs displayed aneuploidy. As these authors note, this rate cannot be applied to all chromosomes (23 × 2%–3% would come to a figure of 46%–69% as the rate for all chromosomes combined, certainly an overestimate); the smaller chromosomes apparently are more prone to erroneous behavior. Different mechanisms could be discerned: classical nondisjunction, predivision (see p. 37), and gonadal mosaicism. The aneuploidy rates correlate with increasing maternal age, most notably in the singlechromatid nondisjunction category (Pellestor et al., 2003). All these data are necessarily influenced by the source of the material: the ova mostly come from women being treated for infertility, who are typically of an older childbearing age, and some of the ova may be of poor quality, although it is of interest that the distribution of abnormality does not differ according to the infertility being due to the female or to the male (Pellestor et al., 2002). What does
seem clear is that in the vicinity of 20%–25% of eggs from this population of women are chromosomally abnormal, about half of these manifesting as full aneuploidy. Maternal age is a most important correlate, and this link is well illustrated in the work of Battaglia et al. (1996; and see Fig. 3–7 in Chapter 3), showing how the structural integrity of the oöcyte’s meiotic apparatus declines as a woman gets older. One particular type of abnormal egg, the giant binucleate oöcyte, is typically diploid (Balakier et al., 2002; Rosenbusch et al., 2002). SPERM
The gamete whose chromosomes are most readily accessible to analysis is the sperm. The earlier sperm karyotyping studies used the “humster” (human sperm + hamster ovum pseudofertilization) test. Guttenbach et al. (1997) reviewed the findings from eight research groups in the field, combining a total of over 20,000 sperm karyotypes from healthy donor men using the humster test. From this accumulated experience, the conclusion was that around 10% of sperm are chromosomally abnormal. Aneuploidy is observed in 1%–3%, and about another 5%–10% of sperm have structural chromosome abnormalities, many of which were presumed to have arisen during spermiogenesis as an immediate postmeiotic event. Fluorescence in situ hybridization (FISH) analysis has enabled chromosome counts to be made on very large numbers of sperm. This approach bypassed any question that the humster test might have selected against abnormal sperm, although in fact it appears that this did not happen (Van Hummelen et al., 1996). Shi and Martin (2000b) reviewed the published experience, observing that over 5,000,000 sperm from about 500 normal men have been analyzed in a number of laboratories around the world, using one-, two-, or three-probe FISH. Considerable variation existed between subjects, probably a biological effect; interlaboratory variation was also noted, presumably reflecting local methodological differences. The average disomy rate for each of the autosomes ranges around the 0.1%–0.2% mark. The figure for chromosome 14 being a little above, at 0.4%, may be artifactual, while higher levels in the G group chromosomes, nos. 21 and 22, are more likely to reflect reality. X+Y disomy is observed in 0.25%. No ethnic differences have come to light, at any rate comparing Caucasians and Chinese (Shi and Martin, 2000a). Neither is there any, or at least
any consistently observed, correlation with paternal age, except possibly with respect to XY disomy. An increased rate of sperm aneuploidy may influence the success of conception, at least as measured at IVF and PGD. A higher rate of sperm abnormality is associated with a lesser rate of success following ICSI, and with a higher rate of aneuploidy in preimplantation embryos (Nicopoullos et al., 2008; Rubio et al., 2009); and consistent with these observations, the outlook for men with increased aneuploidy is improved by preimplantation diagnosis for aneuploidy screening (PGD-AS) (Rodrigo et al., 2010) (see also later section on “Recurrent Implantation Failure”). Fathers of Aneuploid Children. There are very few data on 46,XY men who have actually fathered children with chromosomal abnormalities. Hixon et al. (1998) studied 10 fathers of paternal-error Down syndrome children (the error at meiosis I in three, meiosis II in six, postzygotic in one) and found no differences: these fathers had a mean of 0.15% disomic 21 sperm versus 0.17% in the controls. Similarly, Blanco et al. (1998) studied a group of 15 fathers of children with trisomy 21, and, overall, the fraction of disomic 21 sperm was little different from a control group: 0.31% versus 0.37%. However, of the total of 25 fathers from the two studies, three stood out with twice the level of sperm disomy 21; and two of these also showed an increase in sperm disomic for chromosomes 13 and 22 (Soares et al., 2001a). As for monosomy X Turner syndrome, three-quarters of which may be the consequence of nullisomy X in the sperm, four fathers of TS daughters reported in Martínez-Pasarell et al. (1999) and in Soares et al. (2001b) had increased levels of sperm with a sex chromosome aneuploidy, and also with disomies 13, 21, and 22. From the foregoing, it is tempting to suppose that a minority of normally fertile men may be predisposed to meiotic errors at spermatogenesis, whether generalized or restricted to one chromosome; but the data are as yet too insubstantial to make a firm statement. Whether meiotic recombination occurs may be an important factor influencing the integrity of the disjunctional process (a point well established in female meiosis). Savage et al. (1998) demonstrated that among paternally derived trisomy 21 cases, the rate of chromosome 21 recombination was only about half the expected level. Likewise, Thomas and Hassold (2003) discuss that recombination is very considerably reduced in the pseudoautosomal
Gametogensis and Conception, Pregnancy Loss and Infertility •
379
region (PAR1) of males with 47,XXY Klinefelter syndrome, in whom the causative meiotic error had been shown to be paternal. The conceptions in these cases had been, presumably, 24,XY sperm + 23,X ovum. Shi et al. (2001b) provide corroborative support for this concept in their analysis of sperm from a normal male, this man being usefully heterozygous for two markers in the pseudoautosomal region (DXYS15 and STS). As does any normal man, he produced some disomic 24,XY sperm, among the normal 23,X and 23,Y sperm. Comparing the normal and the disomic sperm in this subject, recombination occurred between the two markers in 38% of the former, and in only 25% of the latter. Men with a Chromosomal Abnormality. Sperm karyotyping studies in men who themselves have an abnormal karyotype, whether balanced (e.g., a reciprocal translocation) or unbalanced (e.g., 47,XYY), are dealt with in the chapters on the particular abnormality. A rather different type of study of the chromosomes—an assessment of their spatial organization within the nucleus—has been applied to translocation carriers; and compared with normal controls, subtle differences in the “packaging” of the chromosomes can be discerned (Wiland et al., 2008) (see also later section on “Factors in the Male”).
Cytogenetics of the Very Early Conceptus An aneuploid gamete (nullisomic or disomic) will lead to an aneuploid conceptus (monosomic or trisomic). A diploid gamete, combining with a normal gamete, will give rise to a triploid conceptus. On simple arithmetic, given that around 20% of oöcytes may be aneuploid and 10% of sperm are abnormal, and simplistically supposing equal fertilizing/fertilizable capacity, the expectation is for about 30% of conceptions to be chromosomally unbalanced. On top of this, dispermy (two sperm fertilizing the one ovum) can cause triploidy. An abnormal postzygotic cell division can give rise to mosaicism, and this may be a common happening (discussed later). A very rare event is the generation of uniparental disomy
due to two gametes being coincidentally nullisomic and disomic for the same chromosome. These several possibilities all add up to a substantial potential for chromosome abnormality in the very early conceptus, in the first days of existence. T HE CL EAVA G E EM B R YO (“ PR EE M B R YO ” ) (DAYS 1 –3 )
The development of the technology of PGD in association with IVF has offered a much clearer view of the frequency of chromosomal abnormalities in the zygote and in the first few days post conception.2 Admittedly, couples presenting for PGD will not, in the main, be a true representation of all couples. One category of patient will, however, be close to “chromosomally typical”: otherwise normally fertile women who are heterozygotes for a mendelian condition, presenting for diagnosis of embryonic sex or for specific mutation testing. These embryos offer the best insight to the true background rate of chromosomal abnormality, with respect to the maternal age groups involved. Pellicer et al. (1999), assessing some or all of chromosomes 13, 16, 18, 21, 22, X and Y, studied 10 mendelian heterozygous women of mean age 34 years, range 30–36. These women had a total of 12 abnormal embryos out of 62 tested (19%), but a considerably higher figure (46%) was observed in a group of older mothers presenting for the same reason (see also Table 23–3). The “atypical” group of patients presenting to the IVF clinic are of course a population of clinical interest, and thus the observations gained from study of them, however unrepresentative they might possibly be of the general population, are very germane to the agenda of the counselor. Numerous studies confirm that a high fraction of unused IVF embryos are karyotypically abnormal, the ranges observed from 30% to 65% (Wilton, 2002). Phenotypically abnormal embryos with multinucleate blastomeres are very likely to be chromosomally abnormal (Kligman et al., 1996). Bahçe et al. (1999), in a study of day 3 embryos, looked at aneuploidies not typically associated with clinical miscarriage or with term pregnancy, and showed that trisomies 1, 15, and 17 occur
2 Strictly speaking, in utero life is divided into three periods: pre-embryonic (the first 2 weeks), up to formation of the primitive streak; embryonic (to the end of the 8th week) when the body forms and organs are constructed; and fetal (from 8 weeks to term), characterized by growth and changes in proportion rather than the appearance of new features. Often the word fetal is used loosely to refer to the entire period, and in IVF parlance (and in the present discussion) the word embryo is routinely applied in reference to the conceptus in the early cleavage stage during the first few days. “Conceptus,” in theory, applies to any stage, but it generally refers to early pregnancy. The conceptus at the one-cell stage—the fertilized egg—is the zygote.
380
•
R E P R O D U C T I V E FA I LU R E
not infrequently. Voullaire et al. (2000b) applied the technique of CGH to the study of embryos that had surpassed the 5-year statutory storage limit in the state of Victoria, Australia, comprising 12 embryos from eight women aged 26–33 years. Upon thawing and culturing, 3 of these 12 embryos were scored as of good morphology, 3 as average, and 6 as poor. Three out of three good-morphology embryos were chromosomally abnormal in one or more blastomeres, and likewise two out of three average-morphology embryos, and four out of six poor-morphology embryos: thus, only a quarter of embryos were normal. Many of the aneuploidies were in mosaic state. Mosaicism of the very early embryo has been one of the more startling discoveries to emerge from the PGD laboratory, although again there is the caveat that it would be unsafe to draw too many conclusions from these laboratory observations concerning the risk for mosaicism in chromosomally normal zygotes, conceived naturally. Many IVF embryos are aneuploid or diploid/haploid mosaics, and even in normal-appearing embryos the fraction, analyzing a limited number of chromosomes, ranges from 17% to 43% (Wilton, 2002). Iwarsson et al. (1999), applying FISH for chromosomes 15, 16, 17, 18, X and Y, found as many as 72% of “good-morphology” freeze-thawed embryos to be chromosomally abnormal, with 57% being diploid mosaics. This mosaicism presumably reflects the fact that the first few postzygotic divisions are particularly susceptible to errors in chromosomal distribution. The main mechanism may be chromosome loss, due to anaphase lag, but chromosome gain is also frequent; mitotic nondisjunction (different cells with the corresponding monosomy and trisomy) occurs in a small minority (Daphnis et al., 2008). When the mosaicism is extensive (several different karyotypes), these embryos may be referred to as being chromosomally “chaotic.” As noted earlier, Voullaire et al. (2000b) showed, using CGH methodology to check on every chromosome, that a majority of surplus embryos of IVF patients were mosaic. A CGH study by Blennow et al. (2001), on embryos from translocation carriers which had been diagnosed as aneuploid on PGD, demonstrated in some that every cell could be different: the absolute maximum mosaicism. THE MO R ULA A N D B LA STOC YST ( DAYS 4 – 5 )
The short period during which the cleavage embryo advances through the morula stage and into the
blastocyst may be an important hurdle during which the development of many chromosomally abnormal pre-embryos arrest, in particular those with extensive mosaicism (Bielanska et al., 2002a). In the largest study of its kind, Fragouli et al. (2008) studied 136 embryos donated for research, of good quality, and which they considered would not differ too greatly from the in vivo situation (although acknowledging that the embryos came from couples undergoing treatment for infertility, and with an average maternal age of 36). They cultured these embryos through to the blastocyst stage. Overall, 39% of these blastocysts were aneuploid, with 35 trisomies and 46 monosomies identified. As expected, the rate was considerably higher in older (37 and over) than in younger women, 48% versus 16%. The simplest state was a blastocyst with a single aneuploidy, with slightly more monosomies (e.g., monosomy 15) than trisomies (e.g., trisomy 19); and the most complicated imbalance was monosomy for six chromosomes and trisomy for five. Only chromosomes 3, 4, 5, and 9 were not represented among the aneuploidies. The number of monosomic blastocysts was an interesting observation: it had previously been considered that the monosomic conceptus could very rarely advance beyond the morula (day 4). The aneuploidy rate was close to the rate expected from the combination of sperm and egg rates, but less than that seen in cleavage-stage embryos, leading these workers to conclude that the abnormalities in the blastocysts were of meiotic origin; and in agreement with Bielanska et al. (2002a) that “chaotically mosaic” cleavage-stage embryos, aneuploid in many or all of their cells due to mitotic errors, will almost all fail at the embryo-morula hurdle. Along similar lines, Daphnis et al. (2008) looked at embryos that had undergone blastomere biopsy and CGH analysis at day 3, cultured until day 5, and then every cell analyzed. None showed a nonmosaic aneuploidy, and thus it could be inferred that the initial zygote had been normal, with all abnormalities having arisen thereafter. From 13 embryos normal at day 3, 10 had become mosaic at day 5, with 6 of these “chaotic.” Of those abnormal at day 3, all remained abnormal, and mostly more so, at day 5, with 70% chaotic mosaic. Trisomy or monosomy of chromosomes 16, 18, and 22 were the most often observed aneuploidies. Partial aneuploidies were also observed; these may be generated at certain fragile sites. The conclusion from this study was that embryos normal at day 3 may retain normality by day 5, but an abnormality at day 3 predicts a poor fate.
Gametogensis and Conception, Pregnancy Loss and Infertility •
381
Mosaicism that arises over the timeframe of days 4–5, but which is not chaotic, and with the existence of a normal euploid lineage, augurs for a progressive normalization of the embryo that can then, in due course, go on to produce a euploid fetus, and a normal child (Fragouli et al., 2008). The abnormal cells fall by the wayside.
THE VER Y EA RLY “EMBRYO PROPE R” ( T HE T H IRD TO TH E SI X T H WE E KS)
The embryo proper, in the sense that the body plan is beginning to be laid out, takes form in the third week, and is barely 1½ mm long; by the end of the sixth week postconception (8 weeks by dates), it will be 1½ cm. This is a timeframe that is not easily studied. First, this appears to be a period during which the threshold for natural abortion is relatively high, and many abnormal embryos seem able to maintain existence (however imperfect that existence might be). Second, the practicalities of collecting intact embryos from very early spontaneous miscarriage, they being scarcely discernible among the products of conception otherwise (chorionic villi the main component), present obvious drawbacks. Nevertheless, Philipp et al. (2003) were able to study by endoscopy the anatomy of embryos in women prior to uterine evacuation for “missed abortion”; in some, growth had arrested, thus enabling a window upon an earlier developmental stage than a standard calculation of gestation-by-dates might have indicated. Some embryos (illustrated in their paper) showed no recognizable external structures (e.g., trisomy 16), while in some an outline embryonic form could be recognized (e.g., trisomies 4, 7). A French group have been able to look at an even earlier stage (and in which growth arrest might not yet have happened), in examining expelled products following very early elective abortion using the drug mifepristone (RU486), following which procedure there is little, if any disruption of the embryonic anatomy (Golzio et al., 2006). One severely malformed embryo (Fig. 23–2), at 25 days postconception age, had failed to develop such crucial organs as the forebrain, the mesonephros (the kidney anlage), or the liver; and the pharyngeal arch system, a fundamental structural framework for the upper body, was absent. (Neither had limb buds developed; but
this is normal at this stage.) The heart was grossly distorted. The chromosomal diagnosis was trisomy 8 (nonmosaic), due to a parental meiotic nondisjunction. It may be that, following this approach, other such embryonic karyotypic-phenotypic correlations will be able to be drawn.
Cytogenetics of Very Early Pregnancy Loss NONI M PL A N TAT I O N A N D O CCULT A BO RT I O N
Although the natural in vivo circumstance might differ from the observations in vitro, nevertheless it is a fair assumption that a substantial fraction of human conceptions have a lethal genetic burden and will not implant. It becomes a semantic question whether the existence of a nonimplanting morula or blastocyst could be described as a pregnancy, and whether its loss could be considered an abortion. Transient implantation may be associated with little or no perturbation of the menstrual cycle, although the woman may fleetingly feel pregnant as a hormonal response is briefly elicited. This is “occult abortion” (Miller et al., 1980). Monosomy, or extensive mosaicism, may be lethal even before the morula converts into the blastocyst, at least on in vitro observation, but some will reach the blastocyst stage, as discussed earlier. Some trisomies impart early lethality. Trisomy 1 may exist in a small fraction of day-3 embryos in an IVF population, and yet it is almost unknown in an established pregnancy (just four recorded cases, presenting as empty sac3; Vičić et al., 2008). Most trisomy 17 is apparently lost in similar circumstances, with a fall in its incidence from 12% at day 3 to only 0.2% in spontaneous abortions (Bahçe et al., 1999; Dunn et al., 2001). The frequency and range of aneuploidies seen in blastocysts is otherwise similar to that seen at the stage of late first-trimester miscarriage, and this is the next major period during which selective pressure is exerted (Cupisti et al., 2003). RECUR R EN T I M PL A N TAT I O N FA I LUR E AT I N V I T R O F ERT I L I ZAT I O N
More than one cause may apply, and a distinction is to be made between maternal (“uterine receptivity”)
3 This expression refers to a conceptus in which a gestational sac and possibly a yolk sac exist, but no recognizable embryonic parts, or at most a “nubbin” of tissue. Blighted ovum and anembryonic miscarriage mean the same thing.
382
•
R E P R O D U C T I V E FA I LU R E
FIGURE 23–2 A very early embryo with nonmosaic trisomy 8 (A, C, and E), due to meiotic I nondisjunction. A normal embryo (B, D, and F) is shown for comparison; each is about 3 weeks post conception, and 3 mm in length. The trisomic embryo is devastatingly malformed. On sagittal section, normal brain structure (the prosencephalon [pros] and rhombencephalon [rh] nicely shown) and regular somite (s) development are clear to see in the normal embryo (F), compared with the gross deformity in the trisomic embryo (E). On ventral view of the trisomic embryo (C), the heart (h1, h2) is bifid, and no liver bud (li in the normal) has formed between it and the anterior intestinal portal (arrowhead). da, dorsal aorta; nt, neural tube; ot, otic vesicle; pa#, pharyngeal arch #; pe, pharyngeal endoderm; tb, tail bud; ys, yolk sac. (From C. Golzio et al., 2006, Cytogenetic and histological features of a human embryo with homogeneous chromosome 8 trisomy, Prenatal Diagnosis 26:1201–1205. Courtesy H. C. Etchevers; reproduced with the permission of Wiley-Blackwell.)
and embryo characteristics. A subgroup of couples undergoing IVF may produce apparently normal embryos, but suffer repeated implantation failure or recurrent pregnancy loss (RIF, RPL) following transfer of embryos to the uterus. Voullaire et al. (2007) compared the frequency of aneuploidy at PGD in embryos from woman with RIF and found a higher rate of complex chromosome abnormality, which they defined as aneuploidy of three or more chromosomes, compared with those who had not. This effect was not related to maternal age. Thus, it may be that, in at least some of these women, there
is an underlying susceptibly for embryos of theirs to undergo chromosomal error during the first two rounds of mitosis. Taking a step back, the gametes from RIF couples may be analyzed. Vialard et al. (2008) analyzed the first polar body of the oocyte and determined an average aneuploidy rate of 35% (range 0–86%). There may be a male factor: these workers also observed sperm aneuploidy rates of 2.1% in the male partner of RIF couples, compared with 0.6% from a comparison group of couples undergoing IVF, where a female factor had been identified as the
Gametogensis and Conception, Pregnancy Loss and Infertility •
383
cause of the infertility. Of 25 couples who had had both sperm and polar body studies, one-third demonstrated no increased chromosomal risk, but in 60%, there was an increased aneuploidy rate in either polar body or sperm, while in 8%, the gametes of both of the RIF partners displayed a chromosomal susceptibility. These observations would suggest that, in two-thirds of RIF couples, recurring meiotic error may be the cause.
Cytogenetics of Spontaneous Abortion and Later Pregnancy Loss CL I NI C AL MIS C ARRI A GE
At the late first-trimester miscarriage, at which stage tissue from “products of conception” is more readily obtainable, we have a clearer idea of how many conceptuses are chromosomally abnormal and what the abnormalities are. Of all recognized pregnancies (recognized in the traditional way, that is), about 10%–15% end in clinical miscarriage—“spontaneous abortion”—mostly toward the end of the first trimester.4 If the products of conception are successfully cultured and karyotyped, in most studies somewhat over a half of abortuses are shown to have a chromosome abnormality (a figure that has increased with increasing expertise and experience, and the application of modern methodologies: Menasha et al., 2005; Zhang et al., 2009c; Rajcan-Separovic et al., 2010). In the case of an empty sac, the fraction with a chromosomal abnormality may be slightly less, about 60%, compared with close to 70% in cases in which embryonic/fetal parts are identifiable on ultrasound (Lathi et al., 2007), and over 80% in those in which multiple external defects are observed through an endoscope prior to operative evacuation (Philipp et al., 2003). Trisomies account for about 60% of all cytogenetic abnormalities identified at spontaneous abortion (Fig. 23–3). The most commonly seen abnormal karyotypes are trisomy 16, monosomy X, and triploidy. Monosomy X and triploidy account for approximately 20% and 15% of all abnormalities, respectively; and as many as 1% of all human
FIGURE 23–3 Chromosomal findings in products of conception from spontaneous abortion. (After Kajii et al., 1980.)
conceptions may have trisomy 16 (Benn, 1998). Double trisomy (trisomy for two chromosomes) is infrequent, this being an observation in 2.2% in one large series (Diego-Alvarez et al., 2006). After aneuploidies and triploidies, structural rearrangements constitute most of the remainder. Molecular methodologies (MLPA and arrayCGH) are powerful, but they may miss most triploidies. SNP arrays have a higher chance of detecting this chromosome state. This was conceptually demonstrated in a recent study based on CVS obtained from women at the time of diagnosis of a missed abortion (Morales et al., 2008). Karyotypes were obtained on 103, and 80% were abnormal. Because 12 were triploidies, the authors argue that semidirect analysis of CVS, which avoids long-term tissue culture, is a low-cost and effective tool to identify chromosome anomalies in miscarriages. The use of SNP arrays and genotyping can, in principle, detect these abnormal ploidy conditions. Zhang et al. (2009c) studied products of conception from 115 spontaneously miscarrying first-trimester pregnancies, applying standard cytogenetics, polymerase chain reaction (PCR)-based microsatellite genotyping, and array-CGH. As expected, cytogenetics
4 The distinction between an embryo and a fetus (and see note 2) in this setting is not necessarily straightforward. Embryonic development may have arrested, and spontaneous abortion will be inevitable, but the pregnancy may continue for one or a few weeks (“missed abortion”), and using apparent gestational age would give a misleading impression. In this case, it is more useful to consider the developmental stage of the embryo in judging the effects of a particular aneuploidy (Philipp et al., 2003). For example, the triploid embryo (not fetus) shown in Figure 23–4 was retained in the uterus until 18 weeks, but development had arrested at the 7–8 week mark.
384
•
R E P R O D U C T I V E FA I LU R E
in those for which culture was successful (80% of cases) revealed a substantial fraction (60%) with aneuploidy, including two with double trisomy. The molecular methodologies detected another 15 cases with abnormality, including some that had been normal on karyotyping, or which had failed to culture. The overall fraction with abnormality barely changed (61%), but the fraction of the whole material with an accurate genomic diagnosis was much increased. Likewise, Rajcan-Separovic et al. (2010) have tested the potential of array-CGH in revealing a greater fraction of imbalance in abnormal embryos with apparently normal karyotypes. FISH analysis of cells in celomic fluid from a “missed abortion” has been proposed as a means to ensure a more accurate cytogenetic diagnosis (Chatzimeletiou et al., 2005). The origin of the abnormality is, in most, an error at maternal meiosis I, and this includes most of the major trisomies: trisomies 13, 14, 16, 21, and 22, with trisomy 18 a possible exception. Robinson et al. (1999) analyzed the originating status of certain of the less studied karyotypes: trisomies for chromosomes 2, 4 through 10, 12, 15, 17, and 20. About three-quarters showed three alleles for the trisomic chromosome, thus confirming a meiotic origin. Most of the remainder are presumed to have been due to somatic errors; some might have been mosaic, but not detected as such. Trisomy 8 is unusual, in that all cases were due to a meiotic error, which stands in contrast to somatic errors being almost entirely the basis of mosaic trisomy 8 that is diagnosed postnatally. Uniparental disomy appears not to be a causative factor in miscarriage (Shaffer et al., 1998; Smith et al., 1998b; Robinson et al., 1999). Phenotypes of the Embryo/Fetus. An embryo or fetus may or may not be identifiable in the products of conception collected at the time of spontaneous abortion due to chromosomal abnormality. Severe growth disorganization can be graded according to whether there is complete absence of any detectable embryonic parts, a tiny nubbin of tissue without recognizable embryonic landmarks, or an embryo in which cephalic and caudal poles can be distinguished (Philipp et al., 2003). The triploid embryo in Figure 23–4 is very deformed but with recognizable face, trunk, and limbs (but not as severely distorted as the trisomy 8 embryo in Fig. 23–2). Seller et al. (2004) describe the only known case of (presumed) nonmosaic trisomy 2, a very severely
FIGURE 23–4 A triploid (69,XXY) embryo. The face has no landmarks other than eyes and a single opening. The anterior trunk is open, with the heart and liver visible. Spontaneous abortion occurred at 18 weeks gestation, but the length is that of 6 to 7 weeks gestation. The disrupted tissue at the neck was the site of biopsy for the cytogenetic analysis.
deformed fetus that survived to 12 weeks gestation. Warburton et al. (1991) provide a graphic catalog of embryonic/fetal phenotypes from their material of about 1300 karyotypically abnormal spontaneous abortuses collected over a 12-year period in New York state, and we have mentioned earlier the illustrations of aneuploid embryos in Philipp et al. (2003). What actually leads to expulsion of the conceptus from the uterus may be the declining vascular and endocrine function of the placental tissue, with decidual necrosis (that is, death of tissue) finally causing uterine irritation and contraction (Rushton, 1981). The underlying process of decline, or at least a contributory factor, may be accelerated apoptosis: Qumsiyeh et al. (2000) observed a higher apoptotic index in villi of the abortus with an abnormal versus a normal karyotype. Twinning. If an abnormal twin dies, the normal twin may ensure continuation of the pregnancy, and only a parchment-like vestige (fetus papyraceus)
Gametogensis and Conception, Pregnancy Loss and Infertility •
385
remains, preserved in the uterus along with the normal twin. A “vanishing twin” has plausibly been proposed in the study of a pregnancy in which two cell lines were identified at CVS, 46,XX and 47,XY,+9. Amniocentesis gave a 46,XX result, and a normal girl was subsequently born. Analysis of a fibrotic area of the placenta gave the same two karyotypes, 46,XX and 47,XY,+9 (Falik-Borenstein et al., 1994). The likely explanation is that a 47,XY,+9 co-conceptus died, and the fibrotic placental tissue was the only remnant. An extremely rare observation is the trisomy that would otherwise lead inevitably to early miscarriage, but in which a monozygous euploid co-twin allows some ongoing in utero survival. These cases may result from a very early postzygotic event that generates a trisomic cell line (or which generates a normal cell line from a trisomic conceptus), and the trisomic co-twin, among other devastating defects, fails to form a heart (“acardius”). The normal euploid co-twin provides the blood circulation to the abnormal fetus (“twin reverse arterial perfusion”). This scenario has been reported with trisomies 2 and 11 (Blaicher et al., 2000), and we have seen one case due to trisomy 3.
detection of chromosome abnormality or allow diagnosis when classic chromosomal analysis has been unsuccessful (Raca et al., 2009). Among liveborn babies, only 1 in 250 has an unbalanced chromosome abnormality on standard karyotyping (see Table 1–3 in Chapter 1). Thus, there has been a very effective selection against those conceptions that were abnormal (Fig. 23–5). RECUR R EN T PR EG N A N CY LOSS 5
Do some couples, themselves karyotypically normal, miscarry on the basis of a predisposition to produce aneuploid conceptions? Ulm (1999) reviews theoretical possibilities through which such a risk might
FE TAL D EATH IN UTE RO, P E RI N ATAL D EATH
Concerning mid-trimester loss, which, coming between miscarriage and stillbirth, may be referred to as fetal death in utero (FDIU) or in utero fetal demise (IUFD), chromosome abnormality may be present in about a half, although at this stage it is the “viable” rather than the invariably lethal aneuploidies that come to light (Howarth et al., 2002). Array methodology enables the detection of subtler imbalances, as Le Caignec et al. (2005) demonstrate in a series of polymalformed fetuses, the pregnancies terminating naturally or due to medical intervention. Similarly, a fraction of pregnancies going through to term, or at any rate to the third trimester, but with the baby stillborn or dying in the early neonatal period (perinatal death), are due to chromosomal abnormality, whether this be full or partial aneuploidy; a common representative of this group is trisomy 18. Again, array-CGH may increase the
FIGURE 23–5 The frequency of chromosome abnormalities at gametogenesis and during pregnancy, demonstrating the effectiveness of selection against aneuploid states. The figures given for gametes through to embryos are very approximate, and considerable individual variation is probable. The oöcyte percentage varies very considerably according to maternal age. The day-3 embryo percentage, drawn from in vitro fertilization data, may exaggerate the true picture in vivo. The figures for fetal and newborn abnormality are quite accurate.
5 Definition from the American Society for Reproductive Medicine (2008): Recurrent pregnancy loss is a disease distinct from infertility, defined by two or more failed pregnancies. When the cause is unknown, each pregnancy loss merits careful review to determine whether specific evaluation may be appropriate. After three or more losses, a thorough evaluation is warranted.
386
•
R E P R O D U C T I V E FA I LU R E
apply: recessive genes, parental chromosome abnormality, gonadal mosaicism, satellite association, and maternal age. A further suggestion, that of maternal skewing of X-inactivation, has since been excluded as a cause (Warburton et al., 2009). A case exists also for the basis of some recurrent aneuploid miscarriage being simply one of randomness, in the setting of a high background rate of aneuploidy in humans, with increasing maternal age the only clear predisposing factor. A common event is common, and not uncommonly it may happen more than once. Addressing this question, Robinson et al. (2001) provided data from a study group of 54 couples having had 2–4 spontaneous known aneuploid/ polyploid abortions, 122 abortions in total. The mean maternal age was 38 years. The distribution and frequency of karyotypes essentially did not differ from the data of a comparison group of 307 single miscarriage cases. Upon stratification for maternal age, a similarity of proportions of karyotypes in recurrent versus single abortions within each age group held up, and likewise upon comparison with an earlier analysis by Hassold et al. (1993). Trisomies in the Robinson study accounted for 80%, and the four most commonly represented abnormalities were trisomies 15, 16, and 21 (14%, 15%, and 8%, respectively) and triploidy (13%). A few couples had a repeat of the same trisomy, but these observations were not made more often than might have been expected by chance. In those cases in which the parental origin of the trisomy could be determined, the great majority, around 90%, were maternal. And yet, although the supposition of randomness and maternal age as the important contributors to recurrent miscarriage is given considerable weight by these data, to the contrary, a small effect indicating a predisposition to aneuploidy recurrence actually was discernible in Warburton’s review of a very large body of prenatal diagnostic data (see p. 285). A somewhat different picture emerged from studies from Pellicer’s group, in which the end-point of observation was FISH analysis of the embryo at PGD, from a patient population having previously experienced multiple miscarriage. In an initial report, nine couples were studied, the women of ages 30–35 years, having had on average four previous miscarriages, and who had presented for IVF with ICSI and PGD as a means to choose chromosomally normal embryos (Pellicer et al., 1999). Of 72 embryos biopsied at day 3 for karyotyping, 66 were analyzable, and in 35 (53%) an abnormality was found using FISH for chromosomes 13, 16, 18, 21, 22, X,
and Y. A control group of similar age (10 women, ages 30–36) had an abnormality rate of only 19%, but a group of six older women (ages 37–41) had a comparable degree of abnormality (46%) to the miscarrying patients. (The controls were drawn from women of presumed normal fertility presenting for PGD for mendelian indications.) The chromosomal abnormalities included nullisomies, monosomies, trisomies, and tetrasomies for various of the five autosomes, and 45,X, 47,XXX and 47,XYY. As with the observations of Robinson et al. earlier, there was no apparent tendency for the same aneuploidy to recur. A tentative conclusion is that a fraction of recurrent abortion may be explained by a predisposition to recurrent aneuploidy, and that this effect is more apparent in younger women. Further work by Pellicer’s group (Rubio et al., 2003) has continued to support these findings, and the figures set out in Table 23–1 show notably higher frequencies of aneuploidies for the tested chromosomes in embryos of those women suffering recurrent miscarriage. The synaptonemal complex gene, SYCP3, appeared a good candidate as a basis for recurrent miscarriage, and Bolor et al., (2009) studied two women, heterozygous for a mutation in SYCP3, each having had three miscarriages, and no normal pregnancies. The case for SYCP3 was, however, not supported in subsequent work from Mizutani et al. (2011). It may seem counterintuitive, but for some couples who have suffered multiple miscarriage, it may be that an aneuploid abortion indicates a better chance for a normal live birth in a subsequent pregnancy than when a miscarriage is euploid (Ogasawara et al., 2000; Carp et al., 2001). Aneuploidy of the abortus may be less often observed in couples who have had a large number of miscarriages, sometimes into double figures, than among those who have had fewer. The probable reason, in such cases, is that a chromosomally normal miscarriage reflected an underlying maternal factor that would apply to all pregnancies, whereas aneuploidy at least offers the hope that better fortune might attend the next ovulation. But this basis is not universally applicable. Bianco et al. (2006), in a preliminary study, showed an increase in the likelihood for an aneuploidy to be found at prenatal diagnosis, in women who had had previous miscarriage, and this risk increased with the number of miscarriages. Couples with Rearrangements. For the small group of people who are heterozygous for a chromosomal rearrangement, pregnancy loss may of course
Gametogensis and Conception, Pregnancy Loss and Infertility •
387
Table 23–1. Frequencies of Embryo Aneuploidies for Certain Chromosomes in a Cohort of Women Having Had Recurrent Miscarriage (467–559 Embryos Analyzed), Compared with a Presumed Normally Fertile Cohort (104–202 Embryos) RECURRENT MISCARRIAGE ANEUPLOID CHROMOSOME OF EMBRYO
13 16 18 21 22 X, Y
<37 YEARS
20 24 10 23 18 11
≥37 YEARS
19 29 12 27 25 12
COMPARISON GROUP <37 YEARS
≥37 YEARS
7 7 5 9 5 7
21 16 15 30 14 15
Notes: The two major classes of aneuploidy were autosomal monosomy and autosomal trisomy. Figures are percentages. Within each clinical category, distinction is made between women below age 37 years and those 37 and over. The rates of aneuploidy for chromosomes 16 and 22 show the most notable differences between the two cohorts, approximately two- to three-fold, across both age groups. For the other autosomes (13, 18, 21), the marked differences are confined to the women <37 years. The comparison cohort comprised women having preimplantation genetic diagnosis for X-linked mendelian conditions. Source: From Rubio et al. (2003).
occur with a much higher frequency, and this briefly stated fact is the basis of much of what is written in this book. In about ½% of couples who have had more than two spontaneous abortions, and in a purely obstetric-gynecologic referral base in which a karyotype is done as a first-up investigation, one or other of the couple is a translocation or inversion carrier (Simpson et al., 1989). Somewhat higher fractions (2%–5%) reported from other studies may reflect a degree of patient selection. These chromosomal rearrangements are typically of sufficient size to be readily detectable at standard karyotyping; and typically of sufficient size, that imbalanced combinations will lead, very frequently, to inevitable miscarriage. An actual example of a chromosomally unbalanced pregnancy leading to spontaneous abortion in the first trimester is shown in Figure 5–10 (in Chapter 5); this was the third miscarriage out of three pregnancies for the couple, the wife being a t(13;16) carrier. Cryptic translocations can also be associated with a risk for miscarriage (Diego-Alvarez et al., 2007). The risk for miscarriage will depend upon the particular characteristics of the rearrangement. As a global figure, for couples, one of whom carries a reciprocal translocation, there is an increased
odds of three to fourfold, compared to chromosomally normal couples who have had repeated pregnancy loss, to have a subsequent further miscarriage (Ozawa et al., 2008). In contrast, the risk for a viable unbalanced form is very low in this group (Barber et al., 2010b). As well as translocations and other autosomal rearrangements, sex chromosome abnormalities may be identified in couples presenting with recurring pregnancy loss. Table 23–2 sets out the karyotypes seen in a Portuguese clinic population (Kiss et al., 2009). While this was a small series, the spread and prevalence of chromosome abnormalities is quite similar to the findings at infertility investigation (cf. Table 23–3).
Cytogenetics of Infertility Infertility is defined as the inability to achieve conception, or the inability to sustain a pregnancy through to live birth (the latter known also as “infecundity”).6 Certainly, it is common, affecting about 15% of couples. It is worth emphasizing that infertility is to be seen in the context of the couple, not necessarily of the individuals separately. An oligospermic
6 Definition from the American Society for Reproductive Medicine (2008): Infertility is a disease, defined by the failure to achieve pregnancy after 12 months or more of regular unprotected intercourse. Earlier evaluation and treatment may be justified based on medical history and physical findings and is warranted after 6 months for women over age 35 years.
388
•
R E P R O D U C T I V E FA I LU R E
Table 23–2. Chromosome Abnormalities in a Series of 108 Couples Having Had Recurrent Pregnancy Loss KARYOTYPE
Sex Chromosomal 45,X/46,XX 47,XXX/46,XX 47,XXY/46,XY 47,XYY/46,XY Autosomal Simple rcp rob inv (pericentric)
NO.
2 1 1 1 3* 2* 1
*One woman carried two rearrangements, a reciprocal translocation, and a Robertsonian translocation. Source: From a Portuguese clinic study 1975–2008; Kiss et al., 2009.
man may be fertile with a “superfertile” female partner, but not with a woman of average fertility, for example. Many causes of infertility exist, involving the male (Skakkebæk et al., 1994) and the female (Healy et al., 1994) partner, and a fraction of these are presumed to be genetically determined (Layman, 2002), and with demonstrable chromosomal causes seen in a minority. Sex chromosomal defects include XXY and XXY/XY in the male, typically presenting with azoöspermia and occasionally severe oligospermia in the nonmosaic state; and Turner syndrome and its variants in the female. The common Yq microdeletion is dealt with later. The XX male and XY female are rare (Chapter 20). Autosomal abnormalities are infrequently seen as a cause of infertility. The reciprocal translocation (especially when an acrocentric is involved) and the inversion may be associated, though infrequently, with severe hypospermatogenesis and moderate to severe oligospermia (Chapters 5 and 9). Robertsonian translocations are occasionally associated with infertility in the male or, less often, the female (Chapter 7). Translocation between a sex chromosome and an autosome is a rarely identified cause of infertility (Chapter 6). Complex rearrangements (Chapter 12) and rings (Chapter 11) typically present an insurmountable obstacle to cell division in the spermatocyte, resulting in azoöspermia; oögenesis is more robust. The frequency of karyotypic abnormality in couples with infertility depends considerably upon the
Table 23–3. Chromosome Abnormalities in a Series of Candidate Couples for Intracytoplasmic Sperm Injection (ICSI), Comprising 2196 Men and 1012 Women (Some Female Partners Not Studied, Since It Had Been Assumed the Infertility Was in the Male) KARYOTYPE
Sex chromosomal* 45,X/46,XY** 47,XXY 47,XXY/46,XY, etc.*** 47,XYY**** Structural Y abn. X or Y rcp with autosome Autosomal Simple rcp Mosaic rcp Complex rcp rob inv (pericentric) Structural abn., unbalanced Total abnormal None of the above Totals
NO. (FEMALE) NO. (MALE)
– – – – – –
8 49 8 8 9 5
7 – – 7 7 –
18 2 2 18 3 4
21 (2.1%) 991 1012
134 (6.1%) 2062 2196
Note: Most, but not necessarily all, of these abnormalities will have been related causally to the infertility. *Excluding a variety of low-level X chromosome mosaicisms in 28 women, of doubtful significance (including them would increase the “Total abnormal” fraction to 4.9%). **Including one 45,X/46,XX/46,XY low-level mosaic. ***Including one 47,XXY/48,XXXY/46,XY mosaic and one low-level 47,XXX/47,XXY/46,XY mosaic. ****Including one 47,XYY/46,XY low-level mosaic. Source: From a France-wide study 1995–1998 (Gekas et al., 2001).
criteria of ascertainment, and quite wide ranges of figures have been produced. Couples presenting to ICSI programs might be supposed typically to manifest a male factor infertility; but van der Ven et al. (1998) were surprised to discover that female partners had about as many chromosomal abnormalities (X aneuploidy, reciprocal and Robertsonian translocations, inversions) as did the males, in a series of 305 couples presenting for ICSI; the experience of Meschede et al. (1998a) was not dissimilar. In an Italian series of 2710 infertile
Gametogensis and Conception, Pregnancy Loss and Infertility •
389
couples, more men were identified with a chromosome abnormality in those who had had ICSI, compared with those having standard IVF, and the least seen in those men in whom simple intrauterine insemination (IUI) was considered appropriate; no such differential applied to the women (Riccaboni et al., 2008). A large French study (Gekas et al., 2001) brought together all the ICSI programs in France over a 3-year period and included some 3208 individuals, 2196 men and 1012 women, who had come forward as candidate couples for ICSI. Gynecologic causes of infertility had been excluded. Each individual had at least 20 metaphases examined. Sex chromosome mosaicism at a level of <10% was categorized as “minor.” In the men, 6% showed a chromosomal abnormality, and in the women (excluding probably insignificant minor sex chromosome mosaicism), 2%. This latter figure may again seem surprising, given that the need for ICSI had been based upon the male partners’ infertility. Certainly, in the fraction of their patients with simple “fertilization failure,” cause unexplained, the proportion of women with a chromosome abnormality was rather greater, at 6.4%; but it remains that 2.6% of the women had an abnormal karyotype even when a basis of the infertility had been determined in the male partner. Support comes from a stringently conducted study in which an age-matched control population had less low-level 46,XX/45,X mosaicism (45,X in at least two cells) than women presenting for an ICSI procedure (Morel et al., 2002b). We return to the comment earlier about infertility being a couple condition. The abnormalities included numerical and structural sex chromosome abnormalities, reciprocal and Robertsonian translocations, inversions, and other structural abnormality (Table 23–3). The French group compared their own data with ten other similar series, and their figures of about 6% and 5% for male and female karyotypic abnormality are close to the averages of about 5% and 4%, respectively. The figure is rather higher (16%) in men presenting with azoöspermia. Considering just autosomal translocations, and in relation to the nature of the infertility, Stern et al. (1999) noted the rate of balanced rcp and rob carriers to be 3% in 219 couples (both partners tested) who had failed more than 10 embryo transfers, and 9% in 130 couples who had 3 or more consecutive first-trimester abortions. (In one couple from the latter group, both were translocation carriers; see Fig. 12–6 in Chapter 12). 390
•
R E P R O D U C T I V E FA I LU R E
A Rare Complexity. A most remarkable coincidence leading to infertility in a young woman is described in Kuechler et al. (2010). Her father was heterozygous for a mutation in the FSHR (follicle stimulating hormone receptor) gene, which is located at 2p16.3; and her mother carried an apparently balanced translocation, t(2;8)(p16.3;p23.1), but which in fact had a microdeletion at the 2p16.3 breakpoint, demonstrable on microarray. This microdeletion removed two exons of the FSHR gene. The daughter inherited this translocation, plus the paternal mutation, and in consequence, the former “unmasked” the latter, no normal FSHR was produced, and folliculogenesis was arrested. FACTO R S I N T H E F EM A L E
Fertility in the 46,XX female begins to fall in the mid-thirties, as the ovarian reserve dwindles. The average maximum number of 300,000 ovarian follicles is reached in midfetal life, at 18–22 weeks gestation, falling to 180,000 by age 13 years, 65,000 by 25, 16,000 by 35, and with less than 1000 remaining at the age of menopause (Wallace and Kelsey, 2010). This fall parallels both the increase in risk for trisomy, and an increasing failure in IVF implantation from the mid-thirties (Spandorfer et al., 2000). An important age-related factor may be a decline in the functional competence of the meiotic spindle, compromising chromosomal distribution and leading to the generation of aneuploid gametes (p. 40; Fig. 3–7). Various sex chromosomal abnormal states, mostly mosaic and containing a 45,X cell line, account for— or are at least associated with—a number of cases of female infertility; autosomal abnormalities are less frequent. Some illustrative karyotypes are listed in Table 23–3. In some, the infertility is primary (there has never been a period of fertility), and in others, secondary (following a previous fertile period). In a Malaysian study, Ten et al. (1990) karyotyped 117 women with primary amenorrhea, who had previously been investigated for other causes, and one third had a sex chromosome abnormality. They were classified as follows: X aneuploidies (8%), X structural abnormalities (7%), presence of a Y (14%), and presence of a gonosomal marker chromosome (2%). Six women were mosaic, all having a 45,X cell line. Secondary infertility may be due to premature ovarian failure (POF), and Devi and Benn (1999) studied 30 women with unexplained secondary amenorrhea under the age of 40 years. Four (13%) had chromosomal abnormalities: an Xq
isochromosome, Turner syndrome mosaicism (45,X/46,XX), an X-Y translocation, and an X-autosome translocation7 (the fragile X premutation as a cause of POF is discussed on p. 248). Failure of the meiotic apparatus, with no formation of the first polar body, may be a rare cause of female infertility, and possibly due to an autosomal recessive gene (Neal et al., 2002; Schmiady and Neitzel, 2002). A Robertsonian translocation may be an uncommon association with recurrent pregnancy loss (Kawano et al., 1998), as may the 47,XXX state (Itu et al., 1990). Kuo (2002) systematically studied 1010 couples having had recurrent loss and found two women with low-grade trisomy 21 mosaicism, each of whom had had at least one trisomy 21 abortus. Older (≥35 years) women who have had three or more miscarriages have an increased risk for a further loss, and trisomy is by far the most probable cause of this, being seen in about 80% of cases (Marquard et al., 2010). In women who have suffered recurrent implantation failure at IVF treatment, in spite of having had a considerable number of embryo transfers, the rate of nonmosaic autosomal translocation carriers in one series of 65 women who had ≥15 failed transfers was 8%, two being sisters with the same translocation (Raziel et al., 2002). (Compare with the 3% translocation figure based upon rather larger numbers, and testing both of the couple, in Stern et al., 1999, noted earlier.) FACTO RS IN TH E MA LE
Fertility is not necessarily synonymous with normospermia and, as mentioned earlier, a man with oligospermia8 may be fertile with a woman of “superfertility” (Krausz and McElreavey, 2001). Nevertheless, much couple infertility is associated with diminished sperm production in the male, and a fraction of this is associated with an abnormal karyotype (Table 23–3) (Gekas et al., 2001). In men presenting with azoöspermia or oligospermia, numerical and structural gonosomal abnormalities (mostly XXY
and Y rearrangements) and structural autosomal abnormalities (mostly reciprocal and Robertsonian translocations) are identified in 3%–13% (De Braekeleer and Dao, 1991; Meschede et al., 1998a; Stuppia et al., 1998; van der Ven et al., 1998; Causio et al., 1999; Dohle et al., 2002; Elghezal et al., 2006). Rare observations include X;autosome translocations, and the small isodicentric 15 and other small marker chromosomes (Eggermann et al., 2002; Ma et al., 2003; Wang et al., 2009d). X Chromosome Abnormalities. The XXY state is the most frequently observed classical karyotype; some of these may be, and more especially in men with extreme oligospermia rather than azoöspermia, very low-level XY/XXY mosaic (Elghezal et al., 2006). Otherwise, mosaicism with a 45,X cell line, 45,X/46,XY, is often associated with infertility (Newberg et al., 1998), as also may be 45,X/47,XYY (Dale et al., 2002), and true XX/XY mosaicism, or chimerism (Sugawara et al., 2005). In some instances, if there is a residual spermatogenesis, artificial reproductive technology might enable fatherhood. The “XX male” is discussed on p. 335. Y Isochromosomes. The typical Y isochromosome, 46,X,i(Y)(p10) or 46,X,i(Y)(q11), removes Yq-located spermatogenesis loci; the condition is seen in nonmosaic and 45,X/46,X,i(Y)(p10) mosaic forms. While abnormal genital and possibly neurodevelopmental phenotypes may be associated with this karyotype (see p. 484), here we are discussing the otherwise normal male presenting with infertility. The typical basis of this may be “Sertoli-only syndrome,” in which the testis lacks germ cells (Lin et al., 2005). While testicular extraction may sometimes enable sperm retrieval in this syndrome, we know of no cases of success in men with a Y isochromosome. We have seen a man, tertiary-educated, with nonmosaic 46,X,i(Y)(p10), in whom attempted testicular aspiration of sperm was unsuccessful (had IVF with ICSI been possible, PGD to select for an XX embryo would have been considered). The isodicentric Y isochromosome with a
7 X-autosome rearrangements associated with ovarian failure may reflect a consequence of breakpoints in one of the Xq21 critical regions (CR1), which bring autosomal “ovarian genes” under the influence of X-heterochromatin at Xq21. In other words, it may be a position effect, whereby autosomal genes for ovarian function are down-regulated due to epigenetic modification (Rizzolio et al., 2006). 8 Oligospermia is defined as a sperm count of <20 million per milliliter. Oligoasthenoteratozoöspermia includes the observations of poor motility (astheno) and an increased fraction of abnormal forms (terato). Severe oligospermia is a count of <2 million per milliliter, moderate oligospermia is 2–5 million per milliliter, and mild is 5–20 million per milliliter. Azoöspermia is absence of sperm. A distinction is to be made between obstructive and nonobstructive azoöspermia; in the latter, the primary fault is a severe defect of spermatogenesis.
Gametogensis and Conception, Pregnancy Loss and Infertility •
391
distal Yp breakpoint (and thus two copies of Yq) is a very rare form, and similarly related to infertility (Codina-Pascual et al., 2004). Yq Microdeletions. The most frequent chromosomal cause of male infertility resides in a Y chromosome microdeletion, with particular reference to the AZF (azoöspermia factor) regions in Yq11, wherein certain spermatogenesis factors have their loci (see Fig. 6–1 in Chapter 6). The fraction varies according to patient selection, and when other causes of oligospermia/azoöspermia have been excluded, the proportion due to AZF deletion reaches 10%–20%. There is a large literature on this subject, reviewed in Foresta et al. (2001). While the initial discovery had been made by cytogeneticists (Tiepolo and Zuffardi, 1976), these Y-deletions are mostly not detectable cytogenetically, and are routinely analyzed using molecular methodology. There are three main AZF regions, a, b and c, and deletions in one or more region can impair spermatogenesis or lead to its complete failure. The most commonly seen deletion involves the AZFc region, in Yq11.23, the causative mechanism being similar to that described in Chapter 19, with inappropriate apposition of duplicons (Kuroda-Kawaguchi et al., 2001). AZFc contains the DAZ—deleted in azoöspermia—multigene family, the products of these duplicated loci being important (but not necessarily crucial) spermatogenesis factors. As a rule, AZFa or AZFb deletions are more severe in their effects than AZFc. Different causes for disordered spermatogenesis may coexist in an individual, and Jaruzelska et al. (2001) point to the need for cytogenetic studies, bringing to attention cases in which 45,X/46,XY mosaicism may have had an additive effect along with an AZFc deletion. A male child conceived from a father with a constitutional Yq microdeletion would very likely have similar infertility (although, as noted later, some men with a Yq deletion may retain fertility). Komori et al. (2001) formally demonstrated that a man with a del(Yq) on blood karyotyping could transmit the deletion, in showing the deletion actually to be present in sperm, as did de Vries et al. (2001) in all of seven infertile men with deletion of the DAZ gene cluster. The observation of the same deletion in the sons of men who had conceived via ICSI confirms the reality of vertical transmission (Cram et al., 2000: Mau Kai et al., 2008). The reduction in fertility may be relative, at least for AZFc deletions, and at a younger age, and perhaps with a partner of 392
•
R E P R O D U C T I V E FA I LU R E
“excellent” fertility, a man with a deletion may father children with no obvious difficulty (Krausz and McElreavy, 2001). Chang et al. (1999) report the example of an azoöspermic 63-year-old man with a DAZ deletion, but who had been fertile in his younger days, having had five children from when he was 25 to 38 years of age. His four sons all had the deletion, and the three of them tested (ages 24–37) were oligospermic or azoöspermic. Translocation Carriers. In the setting of a balanced rearrangement, gametogenesis in the male heterozygote appears more vulnerable than in the female to the complexities imposed by a chromosomal abnormality, and infertility occasionally results. An important element in this male vulnerability may be the integrity at meiosis of the X-Y bivalent, synapsing and recombining at the pseudoautosomal regions at the tips of Xp and Yp—the “sex vesicle” (p. 129). Unpaired or aberrantly associating autosomal segments, particularly of the acrocentric chromosomes, might disturb this integrity, leading to disruption of spermatogenesis (Guichaoua et al., 1990; Oliver-Bonet et al., 2005; Vialard et al., 2006). Another element may be impaired synapsis of homologous segments in the normal and the rearranged chromosomes, which of itself prevents further progress in gametogenesis, and spermatogenesis may be more sensitive to this obstacle than is oögenesis (Hale, 1994; Oliver-Bonet et al., 2005). Pinho et al. (2005) undertook testicular studies on a man with a de novo 46,X,t(Y;1)(q12;q12), demonstrating reduced Xp/Yp pairing, and showing that spermatogenesis had arrested at meiosis I, and that the germ cells had undergone apoptosis. If spermatogenesis is retained, a compromised testicular environment due to the presence of a translocation may nevertheless, of itself, predispose to the production of diploid sperm (Egozcue et al., 2000). Not only may the contained genetic material of the spermatozoön be faulty from the translocation heterozygote but also the “container.” Baccetti et al. (2003) studied by electron microscopy the sperm of an infertile man with a de novo t(10;15)(q26;q12) and showed essentially all sperm to be structurally abnormal. The acrosome was absent or abnormal, cytoplasmic residues were frequent, the mitochondria were often swollen, and axonemal structures were poorly formed. One might have considered that “spermatogenesis genes” on chromosomes 10 and 15 could have been compromised due to being in the monosomic or trisomic state, which then led to these
structural defects in the unbalanced sperm. Yet about a third of sperm had a balanced chromosomal complement. Why were not a third of sperm ultrastructurally correspondingly normal? There is more to be learned about the genetics of spermatogenesis. Notwithstanding the above, it remains true that fertility is usually unimpaired, or scarcely impaired, in male translocation heterozygotes. In their study of just over 10,000 sperm donors, all of proven fertility, the frequency of men carrying reciprocal and Robertsonian translocations, and also pericentric inversions, did not differ with statistical significance from the general population (Ravel et al., 2006a). The semen indices of these men were within normal ranges. This epidemiology indicates that while it is true that a few heterozygotes may have impaired fertility, their numbers are too small to sway the figures of a large carrier population into statistical significance. Sperm Phenotypic Defects Associated with Chromosome Abnormality. A syndrome of infertility associated with “large-headed sperm” is described in Benzacken et al. (2001a). Polyploidy may be the explanation here. Benzacken et al. studied infertile brothers with oligoasthenoteratozoöspermia, half of all sperm having the large-head phenotype. FISH analysis (X, Y, 18) in one brother showed all sperm cells to be diploid or polyploid (3n, 4n, and >4n). The basic fault may lie in a failure of the cell to cleave at the two meiotic stages and, with brothers affected, a genetic cause may reasonably be presumed. Similar cases were reported by Devillard et al. (2002), Lewis-Jones et al. (2003), and Perrin et al. (2008). In the Lewis-Jones study, three men had complete teratozoöspermia (all sperm with abnormal forms, such as double heads, large heads, multiple tails), and the frequency of chromosomal abnormality was very high, up to 100%. Another type of sperm defect is the “tail stump syndrome,” in which the flagellum, the motor apparatus, forms abnormally. Ravel et al. (2006b) report this defect in infertile brothers, both of whom carried a balanced t(5;12). It may be that a “flagellum gene” was disrupted at one of the breakpoints. Normal Karyotype. Among infertile men whose karyotype is normal, and whose sperm count is abnormally low, there is an increase in the sperm aneuploidy/diploidy rate, with the sex chromosomes being the most prone to exhibit disomy (Shi and Martin, 2001). This effect is more apparent in
those men with severe oligospermia, and in those aged 40 and over (Asada et al., 2000). Vegetti et al. (2000) assessed the influence of sperm count and motility, and showed that both these indices correlate with the frequency of sperm disomy, testing chromosomes 13, 18, 21, X and Y. The observations at testicular biopsy in men with severe oligoasthenozoöspermia support this interpretation, with univalents or oligochiasmatic and achiasmatic bivalents frequently being seen (Egozcue et al., 2000). Men with severe sperm indices may have a slight increase in sex chromosome abnormalities in peripheral blood (as a representative somatic tissue), when a very large number (1000) of cells are studied; and this may suggest a more generalized vulnerability of cell division, both meiotic and mitotic (De Palma et al., 2005). As for men with actual azoöspermia, in whom sperm can be obtained only by testicular or epididymal biopsy or aspiration, the early data on fairly small numbers also show elevated disomy rates for some autosomes and the X and Y chromosomes (Martin et al., 2000; Burrello et al., 2002; Mateizel et al., 2002; Palermo et al., 2002; Gianaroli et al., 2005). Mutation in the SYCP3 gene (see later) has been implicated as a cause of spermatogenic arrest (Miyamoto et al., 2003), and other spermatogenesis loci are known (Avenarius et al., 2009). CON S I D ER AT I O N S R EL AT I N G TO I N VI T R O F ERT I L I ZAT I O N
It was reasonable to have imagined that IVFconceived babies might be more likely to suffer a chromosomal abnormality, given the artificial circumstances of their conception, and in particular being aware of the increased rate of abnormality in the sperm of oligospermic men (Pang et al., 1999; Griffin et al., 2003; Silber et al., 2003). But the observation is of little, if any, such risk. One of the largest and most stringent studies addressing this question comes from Australia, in which 6946 IVF babies born in the period 1991–2004 were compared with 20,838 controls (Halliday et al., 2010). The rate of chromosome abnormality in the IVF babies was 0.99%, compared with 0.97% in the nonIVF babies. Among chromosome abnormalities evident at birth (thus excluding some mosaics and sex chromosome imbalances), the respective rates were 0.69% and 0.80%. (Interestingly, the only category of malformation for which a statistically significant increase was discerned for IVF was in respect of “defects of blastogenesis,” comprising a group of
Gametogensis and Conception, Pregnancy Loss and Infertility •
393
typically severe abnormalities which arise in the first 4 weeks post conception.) Other studies have examined the issue of ICSI versus standard IVF, and there is a suggestion that a difference in chromosomal risk, albeit small, might exist. Possibly, the risks for (a) sex chromosomal aneuploidy and (b) de novo structural rearrangement may be increased in the children of severely oligospermic men (who will of course have needed ICSI to achieve conception). In a large French study (Bonduelle et al., 2002), documenting an 11-year experience comprising 1586 ICSI pregnancies in which prenatal diagnosis had been done, de novo structural aberrations and sex chromosome anomalies were seen in 1.6% (cf. 0.5% in the general population). These comprised 10 sex chromosome aneuploidies (XXY, XXX, XYY, and X mosaicisms) and seven structural rearrangements (mostly apparently balanced translocations), along with 8 cases of autosomal aneuploidy (mostly trisomies 18 and 21) All the gonosomal cases involved the father being severely oligospermic, and this male factor, rather than the ICSI procedure of itself, may have been the basis for the increase; the abnormality rate (gonosomal and autosomal) in children of men with sperm counts >20 million per milliliter was only 0.24%. We may conclude that an additional risk of chromosomal abnormality for children conceived from ICSI is marginal and may apply only in the case of men with very low sperm counts. Karyotyping of the oligospermic man is prudently to be done before proceeding to ICSI (Bonduelle et al., 2002; Griffin et al., 2003). Bofinger et al. (1999) provided ICSI to a couple, the husband having severe oligospermia, and the wife being of older childbearing age. At amniocentesis, on the grounds of the mother’s age, a 45,X/46,X,r(Y) chromosome constitution was discovered, and belatedly, the same karyotype was found in the father. The experience of Veld et al. (1997) is equally telling, concerning two men who both, having suffered reproductive misfortune following ICSI, turned out to have a Robertsonian translocation. Epigenetic Effects. Fertilization occurring in vitro is occurring in an artificial environment. It may be that the delicate interplay whereby the epigenetic reprogramming of chromosomes is applied, according to parent of origin, is vulnerable in this artificial 394
•
R E P R O D U C T I V E FA I LU R E
setting (De Rycke et al., 2002); and the question arises that children born from IVF could be at increased risk for an imprinting disorder (Amor and Halliday, 2008). This does indeed appear to be the case with respect to Beckwith-Wiedemann syndrome due to epigenetic error, perhaps more so in the case of ICSI having been employed, and the risk is severalfold that of the general population (Halliday et al., 2004). The case for Angelman syndrome is rather more tenuous, with essentially anecdotal reports by 2008 of just five affected children born, four from ICSI conceptions. Nevertheless, the fact of these three cases all being the category of epigenetic error, and given the rarity of this type, must raise a valid concern. The third imprinting disorder associated with IVF is the “maternal hypomethylation syndrome” (Mackay et al., 2006). Equally, the statistical weight of the thousands of unaffected IVF children is not to be discounted, and this points to a low statistical level of risk.
Genetics and Pathogenesis of Hydatidiform Mole Hydatidiform mole is an abnormal pregnancy that can be considered, in a sense, as a male chromosomal disorder. The pathology and genetics are reviewed in Petignat et al. (2003) and Slim and Mehio (2007). Typically, there is either a completely paternal karyotypic origin (two haploid paternal sets, 2n = 46) or an additional male haploid set (two paternal and one maternal haploid sets, 3n = 69). The presence of two paternal chromosomal complements is referred to as “diandry.” The chorionic villi undergo a degenerative change, forming fluid-filled sacs (hence hydatidiform; mole means mass). The characteristic appearance has long been recognized (Fig. 23–6). The phenotype is marked (“complete mole”) when the genetic origin is completely paternal, and attenuated (“partial mole”) in the presence of a maternal haploid contribution. Most cases are sporadic, but a specific genetic cause in rare recurrent cases may be maternal homozygosity for a predisposing gene. COMPL ET E M O L E
In the complete mole, there is placental tissue— swollen chorionic villi—but typically no embryo identified. The usual karyotype is 46,XX, looking, at first sight, like a normal female karyotype. This is due, in most, to a doubling (endoreduplication) of the chromosomal complement of a single 23,X
FIGURE 23–6. The appearance of hydatidiform mole, probably the first recorded depiction. (From Baillie, 1799.)
sperm, while a minority are dispermic. In either case, there is no maternal chromosomal contribution. With the mole’s nuclear genome being of entirely paternal origin, there is a total uniparental paternal disomy (“uniparental diploidy”). Moles due to doubling of a sperm chromosomal complement are entirely homozygous; in other words, they have a complete uniparental isodisomy. Complete mole occurs more often at the beginning and end of reproductive life in the female: it is more common in the early teenager and in women in their forties (Bagshawe and Lawler, 1982). Some diandric triploid molar pregnancies, when presenting earlier in pregnancy (before 8 weeks), may present a complete molar phenotype, rather than the partial mole usually observed (see later) (Zaragoza et al., 2000).9 A small minority of moles have a biparental diploid genotype, one causative factor of which is apparently maternal homozygosity or heterozygosity for mutation in the NLRP7 gene. This gene has a
role in the acquisition of genomic imprinting as the ovum is produced, and it may also contribute to the ovum’s safeguarding against polyspermy (Deveault et al., 2009; Hayward et al., 2009; Wang et al., 2009a; Slim et al., 2011; Qian et al., 2011). The original explanation was that this endoreduplication of the sperm’s chromosomal complement represented an attempted correction following fertilization of an “effectively empty egg,” that is to say, an egg lacking a viable maternal pronucleus. This construction has been challenged, and Golubovsky (2003) doubts the existence of the empty egg. He proposes instead a schema whereby diploidization (a sort of “triploid rescue”) in very early mitoses follows a dispermic triploid conception and generates 2n cell lines. If it is the maternal complement which is discarded in each cell (perhaps just two cells, following the first mitosis), the genotype is androgenetic: a mole follows. The complete mole typically presents either at early ultrasonography with a “snowstorm” pattern of the placenta on ultrasonography, reflecting the swollen villi, or at first or second trimester vaginal bleeding. There is a widespread and marked hyperplasia of the trophoblast. Where diagnosis is made early, and curettage performed, some nonchorionic elements (yolk sac, capillaries, amnion) may be identifiable, and very occasionally, embryonic parts (Zaragoza et al., 1997; Petignat et al., 2003). Immunostaining for the p57KIP2 protein (the p57KIP2 gene being paternally imprinted) is a useful marker to discriminate between complete (staining-absent) and partial (staining-present) mole (Sebire and Lindsay, 2006). The incidence of complete mole is about 1 in 1500 diagnosed pregnancies, although regional/ethnic variations exist (Slim and Mehio, 2007). In Japan, the incidence has been falling, from about 1 in 400 in the 1970s, to 1 in 650 by the late 1990s (Matsui et al., 2003). There is a small but significant risk of recurrence following one mole, the risk increasing if a woman has had more than one. Recurrences can be of either kind of mole, complete or partial. In a subset of patients, recurrent complete mole is unusual in being diploid and biparental, and as noted earlier,
9 A rare—or rarely recognized—scenario is that of confined placental mosaicism for molar and normal tissue, the infant being normal (Deveault et al., 2009). Photographs of the placenta in one such case give an obvious visual illustration of the mosaicism (Makrydimas et al., 2002). A possible mechanism is that the sperm underwent an inappropriate mitotic division to give two male pronuclei in the zygote, one of which fused with the female pronucleus (to give the cell leading to the normal child), and the other underwent endoreduplication (to produce a cell that gave the molar component). Or this may simply be the end result of chaotic mosaicism in the first few mitoses, with the two surviving cell lines happening one to be normal, and the other androgenetic.
Gametogensis and Conception, Pregnancy Loss and Infertility •
395
the NLRP7 gene has been implicated; we await a full understanding of the mechanism whereby this effect is mediated. PARTIAL MO LE
An additional paternal haploid chromosome set is the basis of most cases of partial mole. This is triploidy, 69,XXX or 69,XXY (rarely 69,XYY), which may typically be the result of a normal ovum fertilized either with two sperm (dispermy) or with a diploid sperm, although other more complex scenarios are proposed (Zaragoza et al., 2000; Rosenbusch, 2008).10 Partial moles typically present as threatened, incomplete, or missed abortion, during the late first or early second trimester, the mean at 12 weeks. There is hydatidiform change of some villi, and the placenta is abnormally large. It is underdiagnosed and may occur in as many as 1 in 700 pregnancies (a figure Jeffers et al., 1993, derive from a review of all 2251 spontaneous abortions occurring in the catchment population of a Dublin hospital over a 3-year period during which there were 19,457 recorded pregnancies). Fetal development, in the very few cases proceeding far enough for this to be assessed, is characterized by a relatively normal growth pattern (McFadden et al., 1993). There appears to be little difference clinically between fetal development in 69,XXX and 69,XXY; the rare 69,XYY form has an earlier lethality (McWeeney et al., 2009). If the triploidy is confined to the placenta, it is possible for the pregnancy to proceed successfully to the birth of a 46,N child (Sarno et al., 1993). Recurrences are on record, and a possible explanation is a genetically determined weakness in the zona pellucida of the ovum, which should act (the “zona reaction”) to prevent more than one sperm penetrating. The double paternal contribution (diandry) is referred to as type I triploidy. Some cases of recurrence might reflect the effect of maternal homozygosity or heterozygosity for the NLRP7 gene, as discussed earlier; this would be consistent with the observation that repeating moles can, in some cases, be of either type, partial or complete. Placental Mesenchymal Dysplasia. A possible clinical confounder, in that it rather resembles
partial mole, but with apparently normal (or nearly so) fetal morphogenesis, is placental mesenchymal dysplasia (Ang et al., 2009). This is a form of mosaicism. The placenta is, in part, normal, and this part has a biparental (and diploid) genotype. The dysplastic part is also diploid, but of paternal uniparental origin. Fetal growth may be affected; a number have been associated with Beckwith-Wiedemann syndrome (Wilson et al., 2008; Jalil et al., 2009). The pathogenetic process, at least in the majority, occurs in the zygote, as the first mitosis gets underway, and lies in a failure of replication of the maternal chromosomal complement, while the paternal complement replicates normally. One paternal complement then joins with the maternal complement to form a biparental cell; the other paternal complement undergoes endoreduplication and produces a uniparental (androgenetic) cell. These two cells then give rise to mosaicism with two lineages, the biparental lineage substantially going on to form the fetus, and the androgenetic lineage responsible for the dysplastic component of the placenta. Intrauterine or neonatal death is a frequent outcome.
GENETIC COUNSELING Recurrent Miscarriage People who have had one or perhaps two miscarriages generally do not come to a genetic clinic, and neither, as a rule, do they have cytogenetic analysis of the products of conception or an analysis of their own karyotypes. Their physician or obstetrician may have advised them that this loss will very likely be part of the 15% or so of all pregnancies that miscarry, and the chance of a successful pregnancy in the future would be good. But having had three miscarriages requires investigation (although others propose testing at an earlier stage; see next paragraph). To use the jargon, such couples have had multiple abortions, recurrent miscarriage, or recurrent pregnancy loss (or to put it in Latin, abortus habitualis). The usual gynecological investigations, and a chromosome analysis of the couple, should be done at this point. If a chromosomal rearrangement is identified, this will probably be the underlying cause (Gadow et al., 1991); but the possibility of a fortuitous discovery is not to be discounted. The precise nature of the
10 Very uncommonly, partial mole has a normal diploid karyotype, with biparental inheritance. One very rare association is with large autosomal trisomy, such as trisomy for almost all of chromosome 4 (Fritz et al., 2000).
396
•
R E P R O D U C T I V E FA I LU R E
rearrangement (consult the appropriate chapter), the reproductive history of any others in the family who have it, and the presence or absence of gynecological pathology allow one to make of judgment of its role in the etiology of the abortions. In the case of recurrent abortion due to a parent being a translocation carrier, Munné et al. (2000b) report that PGD can very substantially reduce the incidence of abortion, and if access to this technology is available, “translocation couples” may wish to consider this option. The majority of couples will have a normal karyotype, 46,XX and 46,XY. In most centers, cytogenetic analysis of abortus material (an expensive and time-consuming procedure) is not routinely done, and so chromosomal normality or abnormality cannot usually be demonstrated. Some have argued that this policy should shift, and Stephenson et al. (2002) speak of “this unfortunate omission” compromising the management of couples presenting with recurrent miscarriage. For women in their late thirties, who have already had miscarriages, trisomy is the major underlying cause; analysis of products of conception can be useful in offering advice to these women, in that the actual cause can now be known, when trisomy is discovered (Marquard et al., 2010). Since the discovery of an aneuploidy can avoid the necessity for further investigation, Stephenson et al. argue that routine karyotyping would actually be cost-effective and have the further benefit of helping couples understand, and thus come to terms with, their reproductive failure, as Sánchez et al. (1999) have also suggested. Jobanputra et al. (2002) propose that a FISH panel for chromosomes 13, 15, 16, 18, 21, 22, X, and Y applied to uncultured abortus tissue would enable a relatively inexpensive screen, with culture proceeding only on those with an apparently normal result; more recently, molecular methodologies have been suggested in this setting (see earlier discussion). A miscarriage due to aneuploidy actually implies a lower risk for miscarriage of a subsequent pregnancy than if the abortus is euploid. However, a previous aneuploid miscarriage may elevate somewhat the risk for subsequent aneuploidy at prenatal diagnosis. IVF with PGD might benefit some women who have had several miscarriages, although this is controversial (p. 429). Lathi et al. (2007) studied the karyotypes from products of conception in women who had miscarried, and they observed that a third of these would have been, in principle, detectable at PGD using 5-probe FISH, and up to threequarters in 9, 10, or 12-probe FISH. Recurrent
implantation failure (which could be seen, in a sense, as a very early form of miscarriage) is often due to chromosomal aneuploidy; in that case, PGD (or polar body analysis in those countries in which PGD-aneuploidy screening is prohibited) may be useful (Vialard et al., 2008) (and see Chapter 26). We have yet to learn how frequently genes such as the SYCP3 synaptonemal complex gene (see earlier) contribute to miscarriage. It may be that mutational analysis of this, and possibly other such genes, will become a part of the investigation of recurrent miscarriage. Women who carry a SYCP3 mutation apparently (although proof is awaited) have a high risk to produce an aneuploid conceptus. F E TA L D EAT H I N UT ER O
Pregnancy loss in mid-trimester is less frequent than in the first trimester, and some may thus see a lower threshold for karyotyping the products of conception. In this case, Howarth et al. (2002) propose offering chorionic villus sampling or amniocentesis, rather than attempting culture of fetal tissue post delivery, in order to improve the chances of getting a definite chromosomal result. WO M EN O F O L D ER CH I L D B EA R I N G A G E
Maternal age is an important factor in recurrent miscarriage. The meiotic apparatus of the oöcyte deteriorates with age; returning to Figure 3–7 (in Chapter 3), the reader can marvel at the disposition of the chromosomes in the eggs of the older women and appreciate how perfectly plausible it is that egg after egg could be aneuploid. The evidence from IVF points to a sharply increasing likelihood for aneuploid conception in women of older childbearing age (Verlinsky et al., 1999). Some instances of reproductive “bad luck” can seem like very bad luck, but, as mentioned earlier, this may still be the true explanation. Ulm (1999) describes her experience dealing with two couples having had several losses, expressing her and their frustration in not being able to provide a satisfactory explanation, nor to offer a precise recurrence risk. In the first case, following a normal child at the mother’s age of 34, the next four pregnancies, in her late thirties, miscarried or were terminated. The first was not tested, and the next three were trisomic 22, 21, and 14, respectively. The sixth pregnancy, at age 40, tested 47,XX,+15 on direct CVS but normal on long-term CVS and at amniocentesis, with a biparental chromosome
Gametogensis and Conception, Pregnancy Loss and Infertility •
397
15 pattern. The infant was developing normally as an 8-month-old. The second case concerned a woman having three miscarriages at age 36–37 years, which karyotyped as trisomy 22, 16, and 21 respectively. We are not much able to ease Ulm’s frustration, other than to point again to the high background rate of human aneuploidy, and to the major influence of maternal age. It is not easy to distinguish, just on the basis of history, which older couples are destined to experience reproductive misfortune. The counselor needs to recognize that many in this situation will go on to have successful pregnancies, but retaining quite some reservation that the risk may be “significant,” and perhaps “substantial,” for women who are getting into their late thirties or forties. For some, with concern that their reproductive years may be limited, IVF with PGD may seem an attractive option, but advice will need to be tempered by the evolving understanding of pregnancy outcomes from PGD for aneuploidy screening in this setting (p. 428).
with nonobstructive azoöspermia and severe oligospermia, but this is unlikely to be the cause in lesser degrees of oligospermia (>5–10 million/ml) (Foresta et al., 2002; Quilter et al., 2003). At present, sperm karyotyping is a discretionary investigation (if available). Karyotyping should be routine in women presenting with primary ovarian dysfunction or recurrent miscarriage. Fragile X premutation analysis should be considered in women with premature ovarian failure (Streuli et al., 2009); a consideration here is the requirement for informed consent, given the other implications of making this diagnosis. Papanikolaou et al. (2005) pose the question, Is chromosome analysis mandatory in the initial investigation of normovulatory women seeking infertility treatment?—and answer in the negative. In other words, and having reviewed over a thousand infertile women with normal ovulatory cycles, the number and type of chromosome abnormalities detected differed scarcely from a normal neonatal population. A greater return came from karyotyping in women with secondary infertility.11
Infertility Infertility is common, and, in Western countries at least, about 15% of couples wishing to have a child are affected (Foresta et al., 2002). Intrinsic fertility cannot be restored in men with persistent azoöspermia associated with seminiferous tubule failure, and neither in women who have had ovarian failure. The counselor will need to understand how disappointing and indeed devastating this may be to some couples (sometimes one of them more than the other), and to be prepared to deal with this. Those for whom assisted reproductive technology may offer hope need to be well aware that this is not necessarily an easy path, and neither can success be guaranteed. Among the catalog of investigative tests that are available, a karyotype is well up on the list. A grouping of experts from the Italian professional community has addressed the question of what tests should be done and when (Foresta et al., 2002). They propose that karyotyping should routinely be done in men with azoöspermia and oligospermia, and in the United Kingdom, karyotyping of men presenting for ICSI is “commonplace” (Griffin et al., 2003). Yq microdeletions should be checked for in men
I NF ERT I L I TY W I T H PA R EN TA L C HR O M OS O M ES A B N O R M A L
If a chromosomal defect is discovered in one or other of the couple, this at least provides an explanation for the infertility and (according to the exact nature of the defect) may prevent the disappointment of undergoing pointless further investigation. In some, artificial reproductive technology may enable a normal/balanced gamete to be identified and retrieved, and used at IVF. Where this is impossible, artificial insemination or IVF using donor gametes offers an entrée to parenthood, and it may enable one of the couple to be a genetic parent. Women. In women with a sex chromosomal abnormality having oöcyte donation, endocrinological management may be necessary to “prime” the reproductive tract (Devroey et al., 1988). But if the internal apparatus is intact, success may well follow, as is rather notably illustrated by the patient reported in Chen et al. (2003b), who had a Turner syndrome variant due to an isodicentric X, and who produced triplets following donation. In some cases
11 Secondary infertility refers to a couple having previously had a successful pregnancy, but who are currently unable to conceive.
398
•
R E P R O D U C T I V E FA I LU R E
the woman’s own mother, with whom of course she shares half her genes, has been the donor. Artificially stimulated ovulation has been attempted in one case of a chromosomal state associated with secondary amenorrhea. Causio et al. (2000) describe a 29-yearold woman with a 46,X,t(X;16) karyotype who had undergone premature ovarian failure, and in whom ovulation was then achieved by treatment with gonadotrophin releasing hormone and follicle stimulating hormone (GnRH and FSH). But no pregnancy resulted. A small fraction of female partners of oligospermic men are found to have low-level sex chromosomal mosaicism. The reality is, according to the experience of Sonntag et al. (2001), that this does not compromise the course and outcome of ICSI. Men. In men with complete spermatogenic arrest, gamete donation may be considered. In those in whom the chromosome defect leads to oligospermia, rather than complete failure of spermatogenesis, IVF with ICSI is a possible means to achieve pregnancy, and PGD will often be appropriate. Otherwise, given the small increased risk for gonosomal aneuploidy following ICSI, a subsequent conventional prenatal diagnosis may appropriately be offered. Translocations and other rearrangements need to be assessed on their merits. A small (but growing) number of cases of fatherhood in men with Klinefelter syndrome have resulted from ICSI (p. 224). Rare sex chromosome abnormalities are judged individually. For example, a sperm study in the case of a man with sex chromosomal mosaicism (45,X/47,XYY), which gave normal findings, was instrumental in a decision not to have preimplantation diagnosis following an ICSI conception (Dale et al., 2002). In the case of a Yq microdeletion, couples choosing the option of IVF with ICSI should know that a male child would be predicted to have, very probably, the same type of infertility (Foresta et al., 2001). Some might consider having PGD to ensure having a daughter; although Kim et al. (1998) comment that “interestingly, after genetic counseling, the decision to proceed with ICSI for the overwhelming majority of couples remains unchanged.” Nap et al. (1999) assessed 28 such couples, and they interviewed the 10 counselors who had seen them, in six clinics in the Netherlands and in Belgium. A considerable majority of couples (79%) chose to continue with plans for ICSI, with only a few choosing donor insemination (7%), or opting out altogether (14%).
I N F ERT I L I TY W I T H PA R EN TA L CHR O M OS O M ES N O R M A L
Where the male has oligospermia, and if IVF with ICSI is to be attempted, there are the grounds (discussed earlier) for presuming a very slightly increased risk for de novo structural aberration or gonosomal aneuploidy. Sperm karyotyping is not routinely practiced as a basis for informing genetic counseling, although this is an area in which research is continuing and opinions are evolving (Griffin et al., 2003; Machev et al., 2005). It may be prudent to offer prenatal diagnosis for an ICSIproduced pregnancy. However, given the immense investment couples will have made to achieve the pregnancy, there may be reservation about proceeding to an invasive prenatal diagnostic procedure, even being aware of a possibly increased genetic risk. In this context, the data from Aytoz et al. (1998) offer reassurance. These Belgian workers compared outcomes in 576 ICSI pregnancies having amniocentesis (singletons) or chorionic villus sampling (twins), with 540 control ICSI singleton and twin pregnancies not having prenatal diagnosis. The fetal loss rates, and certain obstetric indices (preterm delivery, low birth weight), did not differ significantly between the two groups. Further, the data were not tending in the direction of a greater risk for miscarriage in the prenatal diagnosis patients: the odds ratios for fetal loss were 0.86 (amniocentesis) and 0.47 (CVS) compared to the controls. Nevertheless, in a German population, Meschede et al. (1998b) report that only 17% of a cohort of 107 women having undergone ICSI chose subsequent amniocentesis (or fetal blood sampling), the great majority preferring noninvasive ultrasonography or serum screening. This preference was more marked in those who had had genetic counseling prior to entering the ICSI program. In contrast, an Italian clinic recounted a very opposite figure, with 86% choosing invasive prenatal diagnosis (and 100% choosing ultrasound screening); these workers could see “no logical explanation for the great difference” (Monni et al., 1999). The known or suspected risks for an imprinting disorder (Beckwith-Wiedemann syndrome, Angelman syndrome, maternal hypomethylation syndrome, as discussed earlier) in IVF-conceived children raise a question that it may be prudent to advise couples of these concerns, according to the accumulating understanding of these risks. Counselors working in the IVF clinic will want to maintain a watching brief.
Gametogensis and Conception, Pregnancy Loss and Infertility •
399
Chromosomally Abnormal Children Following Pregnancy by Donor Insemination. If a pregnancy achieved through gamete donation turns out to be chromosomally abnormal, should that donor continue to be used? Kuller et al. (2001) surveyed a number of reproductive endocrinologists and obstetrical geneticists to determine the current practice, with particular reference to trisomy 21 and monosomy X. It was clear that no consistent policy was followed. For chromosomal abnormalities generally regarded as being sporadic (or where any predisposition might reside in the recipient rather than the donor), it would seem unnecessary to remove that donor from the panel. I N VI TR O F ERTILIZ AT I ON A N D MULT IP LE P REG N A NCY
Twinning and higher multiple pregnancies are common in IVF, for the obvious reason that often more than one embryo is transferred following IVF, this being a standard policy to improve the chances for a successful implantation. The conservative number of transfers is two, so that if both embryos do implant, no more than twins will result (unless a single embryo might go on to produce monozygous twins). Some clinics transfer more, sometimes for the simple economic reason that if couples can only afford one IVF cycle and transfer, using three (or even more) embryos increases the chance of pregnancy. The disadvantage is, of course, that if most or all of the embryos implant, a high multiple pregnancy results. Whatever might be the risk for aneuploidy,12 that risk will apply to each embryo individually, thus increasing the overall risk that one at least might be chromosomally abnormal. If both abnormal and normal fetuses are present, and diagnosed at subsequent amniocentesis, selective feticide of the chromosomally abnormal fetus may be chosen, or for a lethal aneuploidy (trisomy 13, trisomy 18) the parents may opt to continue the
pregnancy in the expectation that the abnormal fetus will die (Sebire et al., 1997). One of the claims made for PGD is that single embryo transfers can be done with a better expectation of success, and a twin pregnancy avoided.
Hydatidiform Mole The risk of recurrence in a subsequent pregnancy is about 1% (Garrett et al., 2008), and recurrence can be either of the same (complete or partial) or of the other type. Ultrasonographic surveillance is advisable in a future pregnancy. Having had a second mole, the risk for a third is considerably higher, on the order of 20% (Berkowitz et al., 1994). A few of the single cases, but possibly most of the multiple recurrences, may be due to maternal homozygosity or even heterozygosity for mutation in the NLRP7 gene; the counselor will need to consult current literature as we learn more about this gene, and how it acts. These repeating cases typically show biparental inheritance, in contradistinction to the androgenetic basis of the majority of moles. Fisher et al. (2000) suggest that parental origin is worth establishing in those couples who might have been considering IVF with ICSI (to ensure entry of a single sperm), or PGD for ploidy diagnosis, as a means to diminish the risk in a subsequent pregnancy, since such approaches would be futile if the mole(s) had been biparental. A major aspect of management is that the mole may undergo neoplastic transformation (gestational trophoblastic disease). With complete mole, the risk for the development of invasive mole is about 15%, and for the more dangerous choriocarcinoma, it is 3%. The risks are much less with partial mole, the respective figures being 0.5% and 0.1% (Seckl et al., 2000). These facts need to be borne in mind by those women choosing to attempt a further pregnancy. We are unaware of any reports of recurrence of placental mesenchymal dysplasia.
12 If donor eggs are used, it is the age of the donating woman that counts in determining the age-related aneuploidy risk.
400
•
R E P R O D U C T I V E FA I LU R E
PART SEVEN PRENATAL DIAGNOSIS
This page intentionally left blank
24 parental age counseling and screening for fetal trisomy
THE MATERNAL AGE association in Down syndrome (DS) was known long before its chromosomal basis. In 1909, Shuttleworth wrote that “with regard to parentage . . . the outstanding point is the advanced age of the mother at the birth of the child . . . the next point that strikes one is the large proportion of Mongol children that are lastborn, often of a long family” (p. 662). He considered that either age or parity could be an etiologic factor. Subsequently, Penrose (1933, 1934) demonstrated that it was the mother’s age that was the key factor. A powerful insight into the actual nature of the maternal age effect has been afforded by Battaglia et al.’s (1996) study in normal women, showing that the oöcyte’s meiotic apparatus deteriorates with age. Sherman et al. (1994) stated “. . . increasing maternal age is one of the most important factors in
human reproductive failure, as well as being a leading contributor to mental retardation among live-borns.” Hassold et al. (1993) commented that “The association between increasing maternal age and trisomy is arguably the most important etiologic factor in human genetic disease. Nevertheless, we know almost nothing about its basis”; and likewise Wolstenholme and Angell (2000) observed “. . . there is still no consensus of opinion as to how aneuploidy arises in man, and there is a surprising lack of understanding of the basic mechanism(s) of the well-established links to maternal age.” Some suggested factors are outlined in Chapter 3 (p. 39). The maternal age effect in DS—whatever it may be—has been considered to operate upon oögenesis, predisposing to nondisjunction of chromosome 21 predominantly at the first meiotic division.1
1 But note the startling suggestion from Hultén et al. (2008) that maternal gonadal mosaicism may be the important factor; see p. 42.
•
403
In more general terms, segregation of some other chromosomes is vulnerable to the maternal age effect; and, thus, “older women” who are pregnant run an increased risk for having a pregnancy with trisomies 13, 16, and 18, 47,XXX and 47,XXY, as well as trisomy 21. There is also a slight maternal age association with disorders due to uniparental disomy (Ginsburg et al., 2000), this point being discussed in more detail in Chapter 22. Advanced maternal age—or to use the preferable expression of Ginsburg et al. “mothers at advanced childbearing age”—is a common indication for prenatal diagnosis, although becoming less predominant due to the impact of maternal serum and ultrasound screening. Paternal age generally does not usefully enter the equation, at least with respect to autosomal aneuploidies. Fathers of DS children are older than average, but simply because couples are usually of similar ages, a point determined some 75 years ago by Penrose (1934). In a study from the Atlanta and National Down Syndrome Projects, Allen et al. (2009) identified no association with paternal age per se. Concerning gametic studies in older men, numerous sperm analyses have been done, with somewhat conflicting findings (Robbins et al., 1997; Shi and Martin, 2000b; Eskenazi et al., 2002). Some have shown slight increases in some autosomal disomies, and some have shown increases in sex chromosome disomies, with XY disomy being more consistently noted. Other studies report no significant differences in at least autosomal abnormalities comparing older and younger men (one group even using testicular sperm from men in their eighties; Guttenbach et al., 2000). By no means is XY disomy always raised; for example, Luetjens et al. (2002) found, in their study of men under 30 and over 60 years of age drawn from general populations, no differences in XY disomy between the two groups. It is also to be noted that X-Y recombination (its absence being a risk factor for nondisjunction) occurs no less frequently in older than in younger men (Shi et al., 2002). In one study of men who had actually fathered a child with XXY Klinefelter syndrome, the frequencies of 24,XY sperm did increase with increasing age of the father, although it may be an uncommon individual predisposition, rather than an actual age-related factor, that was substantially responsible for this (Lowe et al., 2001; Eskenazi et al., 2002). As for structural rearrangements, a true paternal age effect is considered to exist, albeit at an order of magnitude less than the maternal/aneuploidy association (Sloter et al., 2004). 404
•
P R E N ATA L D I A G N O S I S
Risk Figures According to Maternal Age How old is “older,” and what is “advanced” maternal age at childbearing? Conventionally, the mid to late thirties is taken as the boundary. The risk curve for DS, the major condition of concern, begins to steepen at this period, although there is no sudden jump. Risk figures for individual ages with respect to this and other aneuploidies have been collected in various jurisdictions, and estimates refined according to certain statistical assumptions, and the information from these studies has long been used as the basis of preconceptional and prenatal genetic counseling. These data are also useful in screening programs for fetal trisomy (see later), the woman’s age-related risk being an important datum to be included, along with the various laboratory test results, to derive her overall risk estimate. For trisomies 13, 18, and 21, spontaneous abortion is more likely than for a normal conceptus. Thus, the prevalence of chromosome abnormality is greater at the time of prenatal diagnosis than at term, and we need access to stage-specific figures. Looking through these different windows of observation—at chorion villus sampling (10–11 weeks), at amniocentesis (about 15–17 weeks), at screen-triggered amniocentesis (may be closer to 20 weeks), and at term—the frequency of chromosomal abnormality, for a particular maternal age, progressively reduces. For trisomy 21, it is estimated that about 30% of all pregnancies existing at the time of CVS abort between then and term, and 24% abort during the period from amniocentesis to term (Table 24–1 and Fig. 24–1). Trisomies 13 and 18 (and monosomy X) have high rates of fetal lethality, with the majority of pregnancies aborting. For XXX and XXY, in contrast, there appears to be very little selective loss in the latter part of pregnancy. These matters may be of particular importance to those women who, having had an abnormal result, nevertheless decide to continue a pregnancy. How likely is it that they will have a liveborn baby with the trisomy in question, or that fetal death in utero will supervene? Won et al. (2005) reviewed 392 women who had continued a trisomy 21 pregnancy, and 106 with trisomy 18; the diagnoses had been given somewhat later than might be usual, since these women had entered a public maternal serum screening program at gestations ranging from 15 to 20 weeks, with amniocentesis then offered to those who returned an increased risk result. For trisomy 21,
Table 24–1. Natural Fetal Loss Rates from Early Pregnancy through to Term, Estimated for the Three Major Autosomal Trisomies and X Monosomy ESTIMATED ABORTION RATE (%) CHROMOSOME ABNORMALITY
FROM 10 WEEKS TO BIRTH
FROM 16 WEEKS TO BIRTH
FROM ~20 WEEKS TO BIRTH*
Trisomy 13 Trisomy 18 Trisomy 21 Monosomy X
83 86 31 76
71 74 25 52
32 10
Note: 10 and 16 weeks approximate to the stages at which CVS and amniocentesis are performed. *Trisomy diagnosed at amniocentesis, following population-based maternal serum screening offered at 15–20 weeks. Sources: From Spencer (2001), Snijders et al. (1995), Won et al. (2005), and Savva et al. (2006).
fetal demise occurred in 10%, and for trisomy 18, 31% (Table 24–1). About a third of the trisomy 21 losses happened before the stage of viability (that is, 24 weeks), the comparable figure in trisomy 18 being 15%. In those pregnancies proceeding beyond 24 weeks, the losses were evenly spread according to duration. D OWN SY N D RO ME
The largest body of data to be collated for the agerelated risk of trisomy 21 is that of Morris et al. (2002), who examined records from a 10-year
period, 1989–1998, in England and Wales. We have used their material as the basis of the age-related live-birth figures to age 44 presented in Table 24–2, as probably the best available, although in fact the estimates for younger women (up to age 34 years) have been very similar in all studies, and quite similar in the 35–44 year age bracket (Morris et al., 2003). However, and contrary to earlier interpretations, the risk of having a baby with trisomy 21 does not increase from age 45 and older (Morris et al., 2005a). This might reflect a greater tendency to miscarry an abnormal fetus in women in their late forties and early fifties, or hypothetically a “meiotic robustness”
40 CVS
Prevalence of DS per 1000
35
Amnio
30
Livebirth
25 20 15 10 5 0 36
37
38
39 40 Maternal age (years)
41
42
43
FIGURE 24–1 Prevalence of Down syndrome for maternal ages 36–43, at three “windows of observation”: the time at which CVS is done (about 10 weeks), amniocentesis (15–17 weeks), and at live birth. (From the study of Halliday et al., 1995, courtesy J. L. Halliday.) Parental Age Counseling and Screening for Fetal Trisomy •
405
Table 24–2. Maternal-Age-Specific Risks for Trisomy 21 at Live Birth MATERNAL AGE
PREVALENCE AT LIVE BIRTH
MATERNAL AGE
PREVALENCE AT LIVE BIRTH
YEARS
‰
1 IN
YEARS
‰
1 IN
14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33
0.9 0.4 0.5 0.6 0.6 0.7 0.7 0.7 0.7 0.7 0.7 0.7 0.8 0.8 0.9 1.0 1.0 1.2 1.4 1.7
1108 2434 2013 1599 1789 1440 1441 1409 1465 1346 1396 1383 1187 1235 1147 1002 959 837 695 589
34 35 36 37 38 39 40 41 42 43 44 45 and older
2.3 3.0 3.9 5.0 6.2 8.8 12 15 19 27 26 34
430 338 259 201 162 113 84 69 52 37 38 30
Source: From Table 2 in Morris et al. (2002), the data up to age 44, and from Morris et al. (2005a), for age 45 and older. The figures to age 44 are based on data from just over 6 million births in England and Wales 1989–1998; the figure for 45 and older comes from a review of several sources internationally. Prenatal diagnostic data were included in this material, weighted according to the probability of survival to term. No trisomy 21 pregnancies were recorded at ages 11–13 (274 births) or at ages 53–55 (169 births). The per thousand figures (‰) are rounded.
in some women of this age who are yet able to achieve pregnancy. Estimates for the risks of detection of trisomy 21 at prenatal diagnosis, at the maternal ages at which the procedures would be done, are given in Table 24–3. Systematic calculations from non-Caucasian ethnic groups have come from Nigeria, China, Japan, South America, and Hawaii. Adeyokunnu (1982) showed, in Nigeria, no difference in incidence compared with Europeans; and in a study encompassing nine South American countries, Carothers et al. (2001) demonstrated incidence data and maternal age correlations very similar to those recorded from other jurisdictions. In Japan, Yaegashi et al. (1998) collected data from four clinics, comprising, in all, 5484 pregnancies of women 35 years and older. The risks for trisomy 21 (and for aneuploidies) overall were, on the face of it, somewhat less than in a 406
•
P R E N ATA L D I A G N O S I S
European population. The raw figures did, however, fluctuate somewhat, with rather small numbers of affected fetuses at each age category. A question might be raised whether some cases could have escaped ascertainment by earlier screening and not otherwise recorded. It may be premature to suppose that aneuploidy rates could differ to any important degree between Japanese and other ethnic groups, a view that is supported by the observation otherwise of no significant differences in a Hong Kong population (Lau et al., 1998); but bearing in mind also data from Hawaii suggesting that, at older maternal ages, the DS rate may be somewhat less in a Pacific Island population (Forrester and Merz, 2003b). Taking a more fundamental view, Ghosh et al. (2009) demonstrated that rates of meiotic recombination in families with a DS child in India were essentially the same as in a U.S. population, pointing to a basic
Table 24–3. Maternal-Age-Specific Risks for Trisomy 21 Calculated at 10 Weeks Gestation (the Usual Time for CVS) and at 16 Weeks (Amniocentesis) MATERNAL AGE* YEARS
20 25 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44
GESTATION 10 WEEKS
16 WEEKS
800 710 470 410 350 290 235 185 150 115 90 65 50 40 30 20 15
1050 930 620 540 460 380 310 245 195 150 115 90 70 50 40 30 20
1 in
*Age at the indicated gestation. Source: From Table 2 in Snijders et al. (1995). Figures are rounded.
identicalness of nondisjunctional mechanism across these populations. OT HE R A N EUP LO IDY
The figures for DS are of most interest, as this condition (1) produces a major mental handicap, (2) implies a major burden for parents in that survival well into adult life is now the norm, and (3) is the commonest single chromosome defect in newborns. But the data for other aneuploidies are important. Women seeking advice on their age-related risk and considering prenatal diagnosis should know also that some other rather uncommon trisomies of severe effect (13 and 18) might be detected; and that, on the other hand, there are some age-related sex chromosome aneuploidies (XXX, XXY) that have much milder, but not trivial, effects. Tables 24–4 and 24–5 set out age-related risk estimates for these other categories of aneuploidy. There is also the possibility, irrespective of maternal age, that
some other type of chromosome defect might exist. Table 24–6 sets out the risk for any chromosomal defect, whether maternal-age associated or not, to be detected at prenatal diagnosis. To put these figures into some perspective, we remind the reader that the prevalence of unbalanced chromosomal abnormality in the whole newborn population is approximately 0.5%, or 1 in 200 (see Table 1–3 in Chapter 1). Another window of observation afforded in recent years is at preimplantation diagnosis, and increasing rates of aneuploidy seen in biopsied embryos, according to the mother’s age, are demonstrated in Table 24–7. N O PA R EN TA L A G E EF F ECT I N SOM E D EF ECTS
There is no discernible increasing risk with increasing maternal age for the following chromosomal abnormalities: de novo rearrangement, XYY, triploidy, and unbalanced karyotype due to transmission of parental translocation. For monosomy X, the risk actually lessens with increasing maternal age. With no firmly proven paternal age associations, advanced paternal age is not of itself a particular indication for chromosomal prenatal diagnosis, although a case might hypothetically be made for a much older (sixties and older) father. SECUL A R CH A N G ES I N M AT ER N A L A G E DI ST R I B UT I O N A N D D OW N SYN D R O M E PRE VA L EN CE
Changing maternal age profiles in a population will influence the birth prevalence of DS. In the England of Shakespeare’s time, few women lived long enough to bear children in older age, and along with the effects of poor survival in DS, perhaps no more than 100 individuals with trisomy 21 then existed in that country, in a total population of 4 million (Berg and Korossy, 2001); a similar situation exists in some developing countries today. (Nevertheless, Levitas and Reid, 2003, were able to record a number of probable and possible depictions in art from centuries past, and indeed Martinez-Frias, 2005, has presented a photograph of a terra-cotta head, made in about 500 CE in Mexico, that convincingly captures the essence of the DS facies.) In New Zealand in the 1920s, maternal mortality was much less of an issue but family planning was rudimentary, and about 45% of all mothers were aged 30 and over. The great majority (about 90%) of all DS babies from that
Parental Age Counseling and Screening for Fetal Trisomy •
407
Table 24–4. Maternal-Age-Specific Risks for Trisomies 13 and 18 Calculated at 10 Weeks Gestation (the Usual Yime for CVS), 16 Weeks (Amniocentesis), and at Live Birth MATERNAL AGE* YEARS
20 25 30 35 36 37 38 39 40 41 42 43 44
1 in
TRISOMY 13
TRISOMY 18
10 WEEKS
16 WEEKS
LIVE BIRTH
6500 5600 3700 1500 1200 900 700 530 400 300 230 170 130
11000 9800 6500 2600 2000 1600 1200 920 700 530 400 300 220
14300 12500 11100 5300 4000 3100 2400 1800 1400 1200 970 840 750
1 in
10 WEEKS
16 WEEKS
LIVE BIRTH
2000 1750 1200 470 370 280 220 170 130 95 70 55 40
3600 3200 2100 840 660 510 390 300 230 170 130 95 70
10000 8300 7200 3600 2700 2000 1500 1000 740 530 400 310 250
*Age at the indicated gestation or at birth, respectively. Source: Prenatal data from Tables 3 and 4 in Snijders et al. (1995), and modeled livebirth estimates from Appendix A in Savva et al. (2010). Figures are rounded.
Table 24–5. Maternal-Age-Specific Risks for 47,XXX and 47,XXY at Amniocentesis and at Live Birth MATERNAL AGE
XXX AMNIO
YEARS
33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49
XXY
LIVE BIRTH
‰
‰
1 IN
0.4 0.5 0.7 0.9 1.1 1.4 1.8 2.2 2.8 3.6 4.5 5.7 7 9 11
0.4 0.5 0.5 0.6 0.8 0.9 1.1 1.3 1.6 1.9 2.2 2.7 3.2 3.8 4.5 5.5 6.5
2500 2000 2000 1650 1250 1100 900 770 630 530 450 370 310 260 220 180 150
Source: From data in Tables 20.4 and 20.7 in Hook (1992). Figures are rounded.
AMNIO
LIVE BIRTH
‰
‰
1 IN
0.5 0.6 0.8 1.1 1.4 1.8 2.4 3.1 4.1 5.4 7 9 12 15 20
0.4 0.4 0.6 0.7 0.9 1.1 1.4 1.7 2.2 2.7 3.4 4.3 5.4 6.8 8.5 11 13
2500 2500 1650 1450 1100 900 700 600 450 370 300 230 180 150 120 95 75
Table 24–6. Maternal-Age-Specific Risks for All Unbalanced Chromosomal Abnormalities at Chorionic Villus Samplinga and at Amniocentesis,b for the Age Range 33–45 Years MATERNAL CHORIONIC VILLUS AMNIOCENTESIS AGEC SAMPLING %
33 34 35 36 37 38 39 40 41 42 43 44 45
0.9 1.2 1.5 2.0 2.5 3.5 4.5 6.0 7.5 10 13
1 IN
%
1 IN
115 85 65 50 40 30 22 17 13 10 8
0.5 0.6 0.8 1.0 1.2 1.5 2.0 2.5 3 4 5 6 7
200 160 120 100 80 65 50 40 33 25 20 17 14
a
Including invariably lethal defects Including those for which there is no maternal age effect. c Age at time of procedure. Source: Taken from “averaging” data for ages 33–45 in Tables 20–7 and 20–8 (amniocentesis), and for ages 35–45 from Table 20–10 (CVS) in Hook (1992). Figures are rounded. b
period, at least those surviving to the 1960s to have a chromosome study, were born to mothers in this age group. Over the next four decades, contraceptive practices became gradually more widespread. By the late 1960s, most women were completing their families while still in their twenties, and “older
mothers” made much less contribution to the overall birth rate. Only 20% of all mothers were 30 and over; and the proportion of all DS babies born to this age group had fallen to 53% (Gardner et al., 1973a). We presume, therefore, that the birth prevalence of DS in New Zealand progressively fell over the period 1920–1970. Hook (1992) reviewed the prevalences of DS in various parts of the world during the early 1980s, in relation to the proportions of mothers aged 35 and older. The former Czechoslovakia had the lowest proportion, 3.6%, of older mothers, and Northern Ireland, at 11.1%, the highest. As expected, the observed rates of DS births showed a relationship, with 1.06‰ (per thousand) in Czechoslovakia, and 1.60‰ in Northern Ireland. In the 1980s and 1990s, there was a reversal of the maternal age trend in several parts of the world, with older mothers closing the gap on their younger counterparts. In South Australia, for example, after falling to a trough around 1975–1978, the fraction of mothers over age 35 years progressively rose, and the birth prevalence of DS was anticipated to rise from a low point of about 0.9‰ in the late 1970s to greater than 1.5‰ in 1990–1994 (Staples et al., 1991). In Israel, maternal age dipped in 1978 to a low of 8% of Jewish mothers being 35 or older, and rose to 17% by 1992; and in Alberta, Canada, the comparable figures are 4% in 1980, to 16% in 2007 (Shohat et al., 1995; Lowry et al., 2009). These trends are similar in most affluent countries. Trisomies 13 and 18 have a maternal age association, and so it is not surprising that similar changes in prevalence are observed. From U.K. and Australian data (and adjusting for prenatal diagnosis and termination), the live birth rates increased by 13% and 25% for the two trisomies, respectively, from 1989– 1996 to 1997–2004 (Savva et al., 2010).
Table 24–7. Aneuploidy Rates in 591 Embryos Tested at Preimplantation Genetic Diagnosis in the Course of In Vitro Fertilization, with Respect to Chromosomes 13, 15, 16, 18, 21, 22, X and Y, According to Maternal Age MATERNAL AGE (YEARS)
% Aneuploid % Other abnormal % Normal
25–34
35–37
38–39
40–41
42–44
8 31 61
10 30 60
18 35 47
26 31 43
30 31 39
Source: From Munné et al. (2002b).
Parental Age Counseling and Screening for Fetal Trisomy •
409
The DS birth prevalence is considerably influenced by the use of prenatal diagnosis and selective pregnancy termination, these options becoming widely available in many countries from the 1970s and 1980s. In England and Wales over the period 1974–1987, 14% of potential DS births were avoided by selective abortion, reducing the birth prevalence from 1.26‰ to 1.08‰ (Cuckle et al., 1991). The South Australian figures noted earlier are estimates of the birth prevalences had termination not been used; in fact, the actual prevalences are correspondingly less (Cheffins et al., 2000). More recent data have come from large studies from England and Wales (Morris and Alberman, 2009) and in the United States (Egan et al., 2011), and these reflect the increasing access to noninvasive screening. In England and Wales from 1989 to 2008, while the number of DS diagnoses overall rose very substantially, by 71%, concomitant with a changing maternal age profile, the number of actual DS births fell marginally, from 755 to 743. The proportion choosing termination over this period remained constant, at 92%. In the United States over the period 1989– 2006, the reduction in DS births has varied according to region, with the observed births 44% of expectation in the West, compared with 68% in the Midwest; in the Northeast and the South the figures fell between, but tending more toward those of the West. From their analysis, Egan et al. (2011) conclude that “a Down syndrome fetus is more likely to be prenatally diagnosed and terminated in the West and least likely to be diagnosed and terminated in the Midwest.” The influence of termination is more noticeable among older women: in Alberta, Canada, in 2007, the birth rate in mothers in their forties was 13.2‰, but it would have been 21.5‰ had not termination been available, whereas the comparable rates for 20–24 year-olds were 0.55‰ and a not much greater 0.76‰ (Lowry et al., 2009). Some centers offer a slightly different picture, with the shift to the right in the maternal age curve counteracting the influence of prenatal testing. In Switzerland, the mean maternal age rose from 26 years in 1980 to 30 years in 1996, and the incidence of DS remained practically unchanged (Mutter et al., 2002). Indeed, in Japan, where recourse to termination is less frequently sought, the
birth incidence is rising, as the maternal age spectrum moves to the right (Takeuchi et al., 2008). Prevalence is also influenced by the greater survival of children with DS in recent decades. The survival figure to age 1 year for Western Australia rose from 83% in those born during 1966–1975, to 94% for the period 1991–1996, and survival to age 10 years to 85% (Leonard et al., 2000).
Screening for Fetal Trisomy The birth of a child with Down syndrome is preventable, in that prenatal diagnosis (amniocentesis or chorionic villus sampling) is possible, and a known abnormal pregnancy can be terminated. The definitive prenatal diagnostic procedures of amniocentesis or CVS could not realistically be offered to the whole population of pregnant women,2 but screening allows a subgroup at increased risk to be identified, who then can be offered definitive testing. “Screening” in this context should meet three criteria: it should identify women who are at increased risk, prior to their having a definitive diagnostic test; it should be offered systematically to pregnant women, who are considered only to be at population risk; and it should be seen as beneficial to those who receive it, either in terms of choosing termination, or of being prepared for the birth of a child with DS (Weisz and Rodeck, 2006). The screening tools are the taking of a blood sample, and the performing of an ultrasonogram. Certain biochemical markers in the mother’s serum may have altered concentrations, whether increased or decreased, if she is carrying a trisomic pregnancy; presumably, these differences reflect perturbation in the trisomic feto-placental unit. An assessment is made of the degree to which each level differs from expectation, and these data are factored into an algorithm that takes into account the prior risk due to maternal age (Spencer, 2007). Sophisticated computer packages are employed to calculate an overall risk figure. And, subtle differences in fetal morphology are detectable on ultrasonography. The screening approach differs according to the timing in pregnancy, whether in the first or second trimester (or possibly both: “integrated screening”). If the calculated risk is greater than that of a certain
2 Although the American College of Obstetrics and Gynecology (2007) has advocated that all pregnant women receive genetic counseling and are offered invasive testing. More realistically, screening is becoming widely available, and this College and the American College Medical Genetics recommend that screening be offered to all pregnant women (Skotko et al., 2009).
410
•
P R E N ATA L D I A G N O S I S
threshold risk figure (usually taken as 1 in 250), the pregnancy is regarded as being at “increased risk,” and definitive testing is then offered. Since other aneuploidies can also influence the measured indices, the test procedure in practice becomes broader than just a trisomy 21 screen. If and when the promise were to be realized of definitive prenatal diagnosis of trisomy by the analysis of free fetal DNA in the maternal blood (p. 423)—a form of noninvasive prenatal diagnosis (NIPND)—the screening approach would need to be reassessed. As Wright and Chitty (2009) comment, “It may ultimately be possible to replace the multistep process [of ultrasonography and serum biochemistry] with a single diagnostic blood test based on cell-free fetal DNA or RNA.”3 Even so, it is to be borne in mind that the current screening format picks up other conditions, including chromosomal ones, besides the major trisomies; and thus we would anticipate that screening by maternal serum biomarkers and ultrasonography will have a place in pregnancy management for some time yet. FI RST-TRIMESTER U LT RA SON OGRA PHI C S CRE EN IN G
Ultrasonographic scanning is applied during the window of 11–14 weeks inclusive. This particular parameter is assessed: the degree to which the skin at the neck is separated from the underlying tissue by fluid. Since this fluid does not reflect the sound wave on the scan, it is referred to as “nuchal translucency”; “nuchal thickening” is another expression. An increased nuchal translucency is associated with DS.4 Bekker et al. (2006) propose that the underlying cause is anomalous development of the lymphatic system in the region of the neck; and it appears that this development is susceptible to a chromosomal imbalance. Another ultrasonographic marker of DS is reduced length, or absence, of the
fetal nasal bone; this may be less helpful in the first than in the second trimester (Collado et al., 2005; Ramos-Corpas et al., 2006). F I RST-T R I M EST ER B I O CH EM I CA L SC R EEN I N G
At present, the two first-trimester analytes most commonly measured are β-hCG and pregnancyassociated plasma protein-A (PAPP-A)5, the former typically high and the latter low in a DS pregnancy. We may anticipate that other factors will come to be recognized, and which may improve sensitivity; one such, for example, may be placental growth factor (Cowans et al., 2010). F I RST-T R I M EST ER CO M B I N ED U LT R A S O N O G R A PH Y A N D BI O CH EM I CA L S CR EEN I N G
A better detection is achieved through a combination of first-trimester nuchal translucency assessment and the measurement of maternal serum β-hCG and PAPP-A. If the blood test is done first, these results can be held pending the ultrasound, and the combined figure can be available shortly after the scan is done. Detection rates are typically 80%–90%, for a false-positive rate of 5% or less (Spencer, 2007). The validity of this approach in more precisely targeting an increased risk population is attested in the experience from Denmark, where a national program was put in place in 2004. The number of diagnostic procedures (amniocentesis or CVS) declined from 7524 in 2000 to 3510 in 2006; and yet, over the same period, the number of newborns with Down syndrome fell from around 50 to 30 per year (Ekelund et al., 2008). As the first trimester merges into the second at 12–13 weeks, testing started in one trimester can overlap with the next, and the sensitivity of PAPP-A
3 De Jong et al. (2010) review ethical issues raised by this and other “easy, safe, and early” prenatal diagnostic tests. 4 The sign is not specific for DS, and other chromosomal conditions may also display nuchal translucency. In one very specific example, a subtelomeric deletion of 5q, haplo-insufficiency of a gene FLT4, which is the basis of one type of lymphedema, has been proposed as the cause of the nuchal swelling (Rauch et al., 2003). If the karyotype is normal, there remains a small residual risk for some other type of fetal abnormality, most usually a cardiac defect. The risk is greater for larger nuchal translucency measurements; and the risk diminishes if subsequent mid-trimester fetal ultrasonographic anatomy is normal (Axt-Fliedner et al., 2009). 5 PAPP-A is produced by the syncytiocytotrophoblast, the development of which is impaired in a trisomic pregnancy. The syncytiocytotrophoblast is anatomically close to the maternal uterine vascular circulation, and also to the celomic cavity; the same applies to ADAM12, a more recently developed first- and second-trimester marker (Wang et al., 2010b). Thus, these analytes have value as maternal serum markers (and also in celomic fluid analysis); but due to the barrier imposed by the amniotic membrane, they cannot be applied to amniotic fluid (Makrydimas et al., 2006).
Parental Age Counseling and Screening for Fetal Trisomy •
411
Several “soft signs” on second-trimester ultrasonographic fetal assessment point to an increased likelihood for DS. An advantage is that this procedure is often done routinely, as part of normal obstetric management, and thus a DS screen can be added on essentially at no additional cost. However, the observations do not lend themselves to a ready analysis in terms of adjusting the level of risk; further, the frequency of these “soft signs” in normal fetuses leads to a high false-positive rate. As in first-trimester screening, the length (or absence) of the fetal nasal bone is proposed as a useful adjunctive observation, this measurement being shorter in a trisomic 21 fetus. Ethnic differences in nasal anatomy need to be accounted for (Collado et al., 2005; Jung et al., 2007; Spencer 2007; Casasbuenas et al., 2009).
screening, as high as 94%, for a 5% false-positive rate, the result offered as a single report following the second-trimester testing (Wald et al., 1999). In practice, from a 3½ year study in Hong Kong, Lam et al. (2002) reported an 86% detection rate, which while less than 94% was a better figure than that achieved with either first- or second-trimester screening alone. A U.K. advisory group in 2008 came out against integrated screening as the preferred policy, and in favor of first trimester combined; but this drew a sharply critical response from Rodeck and Ogilvie (2009), who noted the advantages of a lesser rate of invasive testing; a greater use of a more widely available and more accurate procedure (that is, amniocentesis); and more opportunity for natural loss to supervene, with the integrated approach. Okun et al. (2008) provide a useful practical assessment, from their experience in large populations at two major hospitals in Toronto, Canada, in which they could compare first-trimester screening (nuchal translucency plus serum PAPP-A and free ß-hCG) at one hospital, with integrated screening (first-trimester screen followed by second-trimester serum AFP, ß-hCG, and estriol) at the other. Both approaches gave satisfactory detection and falsepositive rates, the integrated approach being (unsurprisingly) slightly superior (88.4% cf. 83.9% detection). These authors point to the need for those who do the nuchal ultrasonography to have “energy and enthusiasm,” in the setting of a busy prenatal clinic. They comment that, as women have become more sophisticated in their understanding of these differing approaches (the study was done over the period 2003–2005), they are well able to make a distinction, and “those most interested in early detection tend to choose first trimester screening, whereas women who wish to minimize the chance of a screenpositive result choose integrated screening.” It was the policy at both hospitals to refer women returning a nuchal translucency measurement of 3.5 mm or above for immediate genetic counseling, given that serum biochemistry, even if normal, is unlikely to bring the risk figure below the threshold for action (thus, a form of contingent screening; see later).
I NT EGRATED S C R EE N I N G
CONT I N G EN T S CR EEN I N G
In theory, the best detection rate could be achieved from a combination of first- and second-trimester
This is a variation on integrated screening, and an example of how this might be applied is given in
can fall away. To counteract this, Jaques et al. (2007) in Victoria, Australia, added in three other analytes (AFP, estriol, and inhibin-A), for those women having the blood sample drawn at 13 weeks, and they referred to this as “augmented screening.” They achieved a 91% detection rate, for a false-positive rate of 4%. SECON D -TR IMESTER BI OC HE M I CA L S CRE EN IN G
The analytes measured in many jurisdictions for second-trimester screening comprise α-fetoprotein (AFP), estriol, the β component of human chorionic gonadotrophin (β-hCG), and inhibin-A (four analytes: hence, the “quadruple test”6). In trisomy 21, the AFP is low, hCG high, uE3 low, and IA high. The proportions of trisomy 21 and other aneuploidies detected by this approach are approximately equal (Sheridan et al., 1997). About a quarter of women aged 37 and older will get an increased risk result, and the remainder a low risk result; as noted earlier, some in this latter group might then choose to forego amniocentesis. SECON D -TR IMESTER ULT RA S O N O G RA P H Y
6 The “double test” uses AFP and estriol; the “triple test” is AFP, estriol, and β-hCG
412
•
P R E N ATA L D I A G N O S I S
Rozenberg et al. (2007). The first-trimester screen is offered, with the expectation of proceeding, in due course, to second-trimester testing. However, a “high-risk” result (taken as greater than 1 in 64) would allow immediate access to a CVS.7 If the screen result were “low-risk” (1 in 1500 or less), no further testing is proposed. With an “intermediate” result (1 in 64–1500), where the majority will lie, the woman continues on track to second-trimester screening, with both results eventually integrated into a final estimation. T WI N P R EG N AN C IES
Two feto-placental units lead, it would be expected, to the production of twice as much of the particular biochemical substance that is then conveyed into the maternal bloodstream. Muller et al. (2003) examined the second-trimester and Spencer et al. (2008) the first-trimester analyte levels in cohorts of twin pregnancies; and the multiple of median (MoM) values were essentially double those of singleton pregnancies. The valid MoMs for risk evaluation can thus be derived by dividing the observed result by approximately 2. Intriguingly, monochorionic (and presumably monozygous) twins at first trimester have a somewhat lower PAPP-A mean (1.76 MoM) than do the dichorionic (2.25 MoM), but with the average (2.12 MoM) being close to double; if chorionicity is distinguishable at ultrasonography, adjustment can be made by applying the appropriate PAPP-A divisor. A theoretical complicating factor, in the case of one (dizygous) twin being trisomic 21, is that the normal co-twin might “dilute out” the abnormal serum biochemistry, and thus invalidate the test result. However, in a large French study addressing a second-trimester population, such an effect, if present, was marginal (and not significant statistically), and screening in this setting was considered to be effective (Garchet-Beaudron et al., 2008). Garchet-Beaudron et al. (2008) point out other issues relating to twin pregnancies. Logically, the age-related risk for DS should double in a dizygous twin pregnancy. But such logic appears not to apply: actual observation does not record such an increase. The technical procedures in the event of an
increased-risk result are more demanding: double amniocentesis, with each sac sampled separately; and, if one twin is trisomic and selective termination is sought, the normal twin is placed at risk. Screening in twin pregnancies requires special expertise. In the case of a “vanishing twin” at the first trimester, as manifest by a second, empty sac, it may be prudent to confine the screening analysis to the nuchal translucency alone (Spencer et al., 2010). I N VI T R O F ERT I L I ZAT I O N PR EG N A N CI ES F OL LOW I N G OVA R I A N ST I M UL AT I O N
The biology of an in vitro fertilization (IVF) conception, following stimulated ovulation with the production of several oöcytes, is subtly different from the natural case. Simplistically, this may reflect the fact of an oöcyte having been released before it was fully prepared to do so; a firmer suggestion is that multiple simultaneous oöcytes generate multiple corpora lutea, which may influence the hormonal milieu, the corpus luteum being a source of hormones, including inhibin-A (Treetampinich et al., 2000). At first trimester, the inhibin-A and β-hCG levels may be marginally elevated, and PAPP-A reduced, compared with the levels from normal conception (the latter two alterations potentially edging up the false-positive rate), although not all studies are in agreement. In the second trimester, changes in levels are slight, but statistically significant: estriol lowered, and β-hCG and inhibin-A raised, but again there is not universal agreement (Lambert-Messerlian et al., 2006; Tul and NovakAntolič, 2006; Weisz and Rodeck, 2006; Anckaert et al., 2008; Amor et al., 2009). The screening analytes appear not to be influenced when the artificial reproductive technology does not involve ovarian stimulation. Couples achieving a pregnancy by IVF may find screening particularly attractive, since it involves no invasive procedure that might put at risk a pregnancy in which there has been so much “investment” (Meschede et al., 1998b). In assessing the degree of risk for aneuploidy, note that it is the age of the woman (whether of the pregnant woman herself, or a donor) at the time the ovum was collected that counts, not her current chronological age.
7 This approach is standard practice in some U.S. centers. If the ultrasound (nuchal translucency, nasal bone) reaches a certain threshold, CVS is offered then and there, while the woman is still on the examination couch. Otherwise, maternal serum testing is subsequently provided.
Parental Age Counseling and Screening for Fetal Trisomy •
413
Interpretation of Screening Results What do these various figures mean? A little epidemiology is in order. Imagine a group of 10,000 pregnant women, of all ages. Assuming a birth prevalence for DS of 1.2‰, we can take it that 12 would otherwise give birth to a baby with DS. If the particular screening approach has a detection rate of, say, 85%, 10/12 of these DS pregnancies would be recognized as being at increased risk, and they could be identified at prenatal diagnosis. The remaining 15% who are carrying a DS fetus (2/12) would fail to be recognized. If the false-positive rate is, say, 4%, 400 women would have an increased-risk report following screening, but followed by a normal result from the amniocentesis or chorionic villus sampling. Putting these figures in the conventional format, we have:
FETUS FETUS NOT TOTAL WITH DS WITH DS
Test shows “increased-risk” Test shows “low-risk” Total
10
400
410
2
9588
9590
12
9988
10000
The detection rate (sensitivity) of the test is 10/12 (~85%). Thus, 15% of women with a trisomic 21 fetus will be missed by the test. The positive predictive value of the test is only 10/410 (2.4%). Thus, 97.6% of women returning an “increased-risk” result will not have a DS baby. The negative predictive value is 9588/9590 (99.98%); in other words, a “low-risk” result means a 99.98% chance for an unaffected baby. The false-positive rate is an important parameter: as noted earlier, this represents the fraction of women who go on to have an invasive definitive test, which then returns a normal chromosomal result. Clearly, the smaller this figure, the better. The tradeoff is this: the smaller the false-positive rate, the less the detection rate. To judge the effectiveness and acceptability of the screening, we can declare a falsepositive rate that is desirable, and this would then determine what the detection rate will be; or we can choose a preferred detection rate and accept the false-positive rate that this would incur. Weisz and Rodeck (2006) summarize as follows. A typically desired false-positive rate is 5%; based upon this, the
414
•
P R E N ATA L D I A G N O S I S
detection rate is noted as DR5. Desired detection rates of 85%, or 90%, would come at a cost of falsepositive rates noted as FPR85 and FPR90. The detection rates, and false-positive rates, that are thus determined, are set out in Table 24–8. T HE UN D ER STA N D I N G O F WO M EN W H O HAVE S CR EEN I N G
The interpretation of a maternal serum test result to the patient is fraught with potential for confusion. The major pitfall is that an “increased-risk” test result may sometimes be understood by the woman and her medical advisor to mean that the pregnancy is likely to be affected. As we showed earlier, the great majority of women who screen “positive” will go on to have a normal baby. Counselors doing this work need a clear awareness of these issues, so that they can enable their patients to understand, intuitively or explicitly, the concept and relevance of a low positive predictive value. The counselor is referred to Macintosh’s (1994) essay “Perception of Risk” for a very readable and practical commentary upon these issues, and to Marteau and Dormandy (2001) for an overview of the complexity of the issues. The ideal is that those having a screening test for DS
Table 24–8. Detection Rates, as Percentages, According to False-Positive Rates, and Vice Versa, due to Four Studies, as Applied to Second-Trimester Maternal Serum Screening
TEST
DR5
FPR85
Triple test (S) Triple test (F) Quadruple test (S) Quadruple test (F)
77 69 83 81
9 14 6 7
POSITIVE PREDICTIVE VALUE
1 in 68 — 1 in 32 1 in 37
DR5 = the detection rate that can be expected for a desired false-positive rate of 5%. FPR85 = the false-positive rate that can be expected for a desired detection rate of 85%. The triple test analytes are α-fetoprotein, estriol, and β-hCG; the quadruple test adds in inhibin-A. S = data from the SURUSS study (Serum, Urine, and Ultrasound Screening Study); F = data from the FASTER study (First and Second Trimester Evaluation of Risks Trial). Source: From Weisz and Rodeck (2006).
should have a basic awareness of the condition, and of the rationale of the screening procedure, and that their beliefs and perceptions and attitudes should be reasonably consonant with the aims and practice of the program. The ideal is not necessarily met. Jaques et al. (2004), in a paper provocatively entitled “Do Women Know That Prenatal Testing Detects Fetuses with Down Syndrome?” surveyed responses from pregnant women 37 years and older in Victoria, Australia in 1998–1999; and the answer to their question was, disconcertingly, that “Down syndrome” was not mentioned as a reason for undergoing pregnancy testing in almost 40% of respondents. Not every woman will respond “rationally” to an increased-risk interpretation, according to the view of rationality as seen by the providers of the screening program. Those who enter into a screening program without being properly aware of the implications may find themselves “in an untenable situation—anxious about a positive result, but unwilling to incur the risks of diagnostic testing” (Kuppermann et al., 2006). Depressive symptoms, and thus a reduced capacity to make clear decisions, may be exacerbated in those with a predisposition, and Hippman et al. (2009) see a role for the counselor in recognizing this. The report from Toronto of more recent date (Okun et al., 2008), in which the program was well received, is rather more encouraging; and equally a subsequent (2003–2004) study from Victoria ( Jaques et al., 2010) suggested that women were responding objectively to risk figures (71% of women chose definitive prenatal diagnosis after an increased-risk screen result, versus only 2.5% after a low-risk result). For those who are better informed, understanding is by no means a neutral matter, and Williams et al. (2005) refer to the role of women as “moral pioneers,” in coming to terms with the ethical issues that readily available screening may, in these modern times, present. Susanne et al. (2006) assessed responses in women who had had what turned out to be a false-positive screening result, following them prospectively through the pregnancy and after the baby was born. Several declared that they had “withheld” their pregnancy, and only returned to reacceptance after the normal chromosomal result from amniocentesis had been conveyed; nevertheless, most would have
the same testing in a future pregnancy. Counselors need to be well attuned to these several complexities; and if a woman’s family physician can share in the decision-making process, this is typically well received (Légaré et al., 2011). Concerning the facts about DS itself in the context of pregnancy screening, leaflets are the simplest means of conveying information, and many clinics/ jurisdictions produce their own material, the quality of which may vary considerably (Murray et al., 2001). Videotapes may be helpful (Hewison et al., 2001). It is a fine matter to judge what should be the level and tone of the information. Bryant et al. (2001) reviewed the leaflets produced in a number of clinics in the United Kingdom and considered that the viewpoints expressed were, in the main, weighted unduly negatively toward DS. It is true that information ought to be couched in such terms that it will be useful, in the fullest sense of that word, to the wide range of people for whom it is intended (and see p. 16). Equally, the comment can be made that attempting to neutralize negative aspects of DS may send a mixed message, since being given the option of abortion in order to avoid having a DS child rather plainly implies that having such a child may not be a desirable outcome. The view that is offered should be clear, accurate, and even-handed.8 E F FECT O F S CR EEN I N G UPO N PRE VA L EN CE O F D OW N SYN D R O M E
In several centers, the increasing acceptance of maternal serum screening has been associated, as expected, with a reduction in the number of DS babies being born, above that achieved by maternal age-targeted amniocentesis. Cheffins et al. (2000) assessed the situation in South Australia. The fraction of pregnant women having screening rose from 17% at the time of its introduction in 1991 to 76% by 1996; in consequence, the proportion having amniocentesis rose from 6% to 10%, with younger mothers the main contributor to this rise. The resulting increase in the detection of fetal trisomy 21, and with termination of the affected pregnancies, caused the birth prevalence of DS to fall by more than half—by 60%, to be precise—from 1.05 to 0.42 per 1000 births. This fall took place in spite of a “natural” increase in prevalence due to the mothers in 1996
8 These matters are dealt with in considerable detail in a document from the National Health Service of the United Kingdom, “Psychological Aspects of Genetic Screening of Pregnant Women and Newborns: A Systematic Review” (Green et al., 2004).
Parental Age Counseling and Screening for Fetal Trisomy •
415
being older. A very similar picture is seen in Belgium (Verloes et al., 2001). During 1984–1989, 244 cases of trisomy 21 were diagnosed in the Genetic Centers of Liège and Loverval, 17% of these at prenatal diagnosis. A decade later (1993–1998), of the 294 diagnoses of trisomy 21, the fraction detected prenatally had risen to 56%, and over 90% of these pregnancies had been terminated. Theoretically (and very probably actually), this reduced the birth prevalence from 1.26 to 0.62 per 1000, a fall of just over a half. More broadly, the pickup rate at prenatal diagnosis for all aneuploidies has increased, due to the identification of increased risk pregnancies afforded by these screening programs. If screening is not widely offered, an inappropriate section of the population may be targeted for prenatal diagnosis, and the reduction that is achieved in DS incidence will be less. In Denmark over the period 1980–1998, there was a rather high level of CVS and amniocentesis procedures being done, but mostly in lower risk pregnancies, and this yielded about a 30%–40% reduction in the expected DS incidence. This figure could have been higher, for a lesser number of invasive procedures, had screening allowed a preselection of women at increased risk (Larsen et al., 2001). P RE VIO US P REG N A N CY W I T H FAL SE- P OS ITIV E RESU LT
If there has been a false-positive test result from second-trimester maternal serum screening in one pregnancy, there is a considerable likelihood, about 20%, that the same thing could happen in a subsequent pregnancy. Naturally, maternal factors influence the process whereby analytes produced by the fetus and placenta are transferred to the maternal bloodstream, and it is unsurprising that these same factors might obtain in sequential pregnancies. This phenomenon can readily be taken into account in a following pregnancy, by applying to the observed MoM, a divisor derived from the MoM values that had been seen in the initial pregnancy (Wald et al., 2004). OT HE R A B N O R MA LI T I ES
Trisomies 13 and 18, Rare Trisomies, Other Aneuploid States. While the prime focus of screening is on trisomy 21, a side benefit is the detection of other, and typically more severe chromosomal disorders. Trisomy 13 and trisomy 18 both show reduced levels of β-hCG and PAPP-A at first-trimester 416
•
P R E N ATA L D I A G N O S I S
screening, more so in trisomy 18, along with increased nuchal translucency or frank cystic hygroma; and using a trisomy 21 analytical algorithm, about three-quarters of these trisomies can be detected, for a false-positive rate of 3%–6% (Breathnach et al., 2007; Kagan et al., 2008b). If specific algorithms for trisomies 13 and 18 are applied, and if an assessment of fetal heart rate is factored in, detection reaches 95%. Few other trisomic pregnancies proceed through to the time of screening. Unsurprisingly, those that do, display abnormalities at screening. For example, in trisomy 22 at firsttrimester screening, the β-hCG is very elevated, PAPP-A somewhat reduced, and fetal growth restriction is typical (Sifakis et al., 2008). In nonmosaic trisomy 9, the biochemistry is similar to that of trisomy 18 (Priola et al., 2007), and the same may apply in the mosaic case. An increased nuchal translucency can be associated with a number of rare aneuploid states, partial or complete (Chen et al., 2005a). For example, β-hCG can be very high in fetal cri du chat syndrome (Torun et al., 2009), and similarly in confined placental mosaic trisomy 16, in which AFP is also elevated and PAPP-A low (Neiswanger et al., 2006; Petracchi et al., 2009). The 22q11 deletion syndrome, of which a heart defect is a frequent component, might have seemed a useful candidate for testing in the context of an increased nuchal fold; but in a smallish study of 146 pregnancies with a nuchal translucency above the 99th centile and otherwise normal, Lautrup et al. (2008) identified none with this deletion. At secondtrimester quadruple-test screening, about two-thirds of non-DS aneuploidies are detected, for a falsepositive rate of 9% (Breathnach et al., 2007). The fact remains, however, that about half of all fetal chromosomal abnormalities, and which would lead to substantial phenotypic defect, are not detected by these noninvasive tests (Grati et al., 2010). Triploidy. The biochemical indices at firsttrimester screening in a triploid pregnancy are quite abnormal, and very differently so according to the category of triploidy, digynic, or diandric (p. 287). In the former, β-hCG and PAPP-A are both much reduced, while in the diandric type, β-hCG is greatly elevated, and PAPP-A marginally reduced. Likewise, ultrasonography is distinctly different, with severe growth restriction in the digynic type, and nearer normal growth but with an enlarged and partially molar placenta in diandric triploidy (Kagan et al., 2008a).
25 prenatal diagnostic procedures
THE MEANS to diagnose the fetal karyotype have provided medical cytogenetics with one of its major areas of application. The discovery of an abnormality allows the option of termination of the pregnancy or, later in gestation, a more suitable obstetric management. The main indications for prenatal cytogenetic diagnosis are the following: (1) the pregnant woman being of advanced childbearing age, (2) parental heterozygosity for a chromosome rearrangement, (3) the birth of a previous child with a chromosome defect, (4) abnormal maternal blood biochemistry, and (5) fetal anomaly detected on ultrasonography.
FETAL ULTRASONOGRAPHIC ANOMALIES Mid-trimester ultrasonographic screening is a routine part of obstetric management, and the identification of a fetal malformation is a common indication for a chromosome study. In Victoria, Australia, for example, 20% of prenatal chromosome tests in 1999 were done on the grounds of ultrasound findings of
a fetal malformation, or of a marker of aneuploidy (Webley and Halliday 2001). Certain major ultrasonographic defects are fairly specific: for example, holoprosencephaly predicts the likelihood of trisomy 13, fetal hydrops/cystic hygroma predicts monosomy X or trisomy 21, an endocardial cushion defect or duodenal atresia predicts trisomy 21, and conotruncal defects are associated with the 22q11.2 deletion. Certain renal defects have a frequent association with fetal aneuploidy, as do cardiac malformations generally (Amor et al., 2003; Wimalasundera and Gardiner, 2004; Carbone et al., 2011). Up to 33% of heart defects are associated with fetal aneuploidy, although in most there will be additional anomalies. Cysts of the choroid plexus (tissue within the cerebral ventricles) are a “soft marker” for trisomy 18, but not trisomy 21 (Walkinshaw, 2000); they are harmless in normal children (DiPietro et al., 2011). On the specific question of rare autosomal abnormalities (rare trisomies, deletions, duplications, supernumerary markers, various other structural •
417
rearrangements), a large European series based upon reports from malformation registers in several jurisdictions linked ultrasound findings to cytogenetic results (Baena et al., 2003). Nearly half of all rare autosomal abnormalities showed fetal anomalies on ultrasonography, with heart and brain defects and growth retardation more often seen with deletions, and cystic hygroma, hydrops, and nuchal translucency more typically associated with trisomies and duplications. These rare abnormalities comprised 7% of all chromosomally abnormal prenatal diagnoses. A specific observation, such as aortic narrowing or a conotruncal defect, may allow a targeted cytogenetic study, such as FISH for the 7q11.23 deletion of Williams syndrome, and the 22q11 deletion (Krzeminska et al., 2009). Asymmetrical growth (head circumference vs. crown-rump length) may point to triploidy (Salomon et al., 2005). The acardiac fetus is often due to an otherwise unsurvivable autosomal trisomy, possibly tempered by mosaicism with a normal cell line, but their existence being maintained by a (karyotypically normal) monozygous co-twin (the “pump twin”). In which cases should a chromosome analysis be conducted, following the discovery of structural anomalies by ultrasound examination? Staebler et al. (2005) examined the karyotypes (on classical cytogenetics) in 428 fetuses with ultrasounddetected anomalies over a 10-year period. The karyotype was abnormal in 9% of cases with an isolated malformation, and in 19% of cases with multiple malformations. The following isolated defects were typically associated with a normal karyotype: hydronephrosis with high obstruction, unilateral multicystic dysplastic kidney, gastroschisis, intestinal dilatation, cystic adenomatoid malformation, pulmonary sequestration, tumor, and vertebral anomaly. Thus, one of these as a single malformation is not an indication, whereas, clearly enough, the presence of multiple malformations would warrant chromosome study. Applying the more precise tool of array-CGH, Coppinger et al. (2009) and Kleeman et al. (2009) found that 2%–4% of pregnancies with abnormal ultrasound findings and a normal karyotype have a clinically relevant DNA copy number alteration. Although the number of cases was too small to stratify the data based on malformation type, these results suggest that all fetuses with ultrasound abnormalities might warrant a microarray analysis; and the American College of Obstetricians and Gynecologists (2009) has recommended that targeted microarray analysis be offered as an adjunct 418
•
P R E N ATA L D I A G N O S I S
in prenatal cases with structural anomalies and a normal karyotype. The minor marker of “increased nuchal fold thickness” (actually, this separation of the skin from the underlying tissue can extend from as far as the occiput down to the lower back) indicates an increased risk for fetal aneuploidy, the level of risk proportional to the degree of separation, and this observation is sufficiently robust that is used for pregnancy screening of aneuploidy, and especially during the first trimester (Chapter 24). This is particularly sensitive in trisomy 18, with the great majority, 90%, manifesting this sign in the first trimester (Sepulveda et al., 2010). Daniel et al. (2003a) reviewed 1800 cases in which an anomaly (an actual malformation, or a minor marker of aneuploidy) had been detected at ultrasonography, and assembled a table of risks of aneuploidy according to the findings (Table 25–1). The abnormal karyotypes included trisomies 13, 18, 21, triploidy, 45,X and mosaics, various autosomal and gonosomal duplications and deletions, rare trisomies, and de novo apparently balanced rearrangements. Souka et al. (2005) have assessed risks in the setting of the specific finding of an increased nuchal translucency, related to the degree of separation (Table 25–2). A practical question is this: if, following the observation of an increased nuchal translucency a CVS or amniocentesis is done, and the chromosomes are normal, is there a residual risk for some other type of fetal abnormality? If the translucency resolves, and if no defects (with particular focus on the fetal heart) are seen at 14–16 weeks gestation, the prognosis is good, with a better than 95% chance of a baby with no major abnormalities; and if a further scan at 20–22 weeks is normal, the risk is likely to be no different than the background population figure. If, on the other hand, the increased translucency persists, a possibility exists of a congenital infection or a genetic syndrome. There are some subtleties in the choice of language when fetal anomalies are uncovered by ultrasound, as de Crespigny et al. (1996, 1999) discuss. We speak of the pregnant woman as a mother, yet she is not; neither is her husband/partner as yet a father. Equally, the fetus is not a baby, not acquiring that status until ex utero existence is achieved. But of course many parents-to-be, not to mention professionals (including us), use these words. Counselors should be sensitive to these subtleties. De Crespigny observes that, if an ultrasonologist should discover a fetal defect, using the terms “baby” and “mother”
Table 25–1. The Likelihood of Discovering a Chromosome Abnormality at Prenatal Diagnosis, According to the Pattern of Defects Identified at Fetal Ultrasonography, for All Maternal Ages LIKELIHOOD OF AN ANEUPLOIDY
DEFECTS
CNS/cranial shape plus cardiac* Key malformation,** singly or in combination CNS ± other*** Nuchal translucency, first trimester ± other abnormality Nuchal translucency, second trimester ± other abnormality Cardiac ± other abnormality Pyelectasis/two vessel cord/ echogenic bowel/short femur Other (singly or in combination)
53% 37%
may exert indirect pressure on the couple to continue the pregnancy: “Although many women regard a fetus as a baby from the very beginning, others will be affronted if their doctor does not seem to recognize this difference between a fetus and a baby, which they may interpret as interfering with the pregnant woman’s reproductive freedom.” As always, counselors will need to know their patients, and to judge the right words to use, and the way to say them (Benkendorf et al., 2001). Twins In the event of a twin pregnancy having been shown on ultrasonography, the question arises of an appropriate prenatal diagnostic procedure, if this is considered appropriate. A point to make here is that, while monozygous (MZ) twins would be expected to have the same karyotype, this does not invariably apply. Furthermore, the ability to interpret monozygosity is not perfect; and those MZ twins in which the split occurred shortly after conception, prior to the differentiation of the extrafetal tissues, may have the same ultrasound morphology of membranes (amnion and chorion) as would a dizygous pair. Thus, the advice is that dual amniocenteses, rather than CVS, may be the procedure of choice; and more especially so in the setting of discordance for an anatomical anomaly (Lewi et al., 2006).
21% 25% 13% 9% 6% 3%
Notes: Some percentages are considerably higher or lower for older and younger maternal ages, respectively. These data were obtained prospectively. *Excluding anencephaly/spina bifida. **Cystic hygroma/hydrops/exomphalos/severe IUGR/duodenal atresia/talipes. ***Excluding anencephaly/spina bifida/cardiac, including choroid plexus cysts. CNS, central nervous system; IUGR: intra-uterine growth retardation. Source: From Daniel et al. (2003a).
PRENATAL LABORATORY DIAGNOSTIC PROCEDURES Since the early 1970s, prenatal diagnosis (PND) of chromosome disorders has been done by culture of
Table 25–2. Likelihood of Chromosome Abnormality, and of Other Outcomes, in the Setting of Increased Nuchal Translucency NUCHAL TRANSLUCENCY
<95th centile 95–99th centile 3.5–4.4 mm 4.5–5.4 mm 5.5–6.4 mm >6.5 mm
CHROMOSOME ABNORMALITY
0.2% 3.7% 21% 33% 51% 65%
MISCARRIAGE, FETAL DEATH
1.3% 1.3% 2.7% 3.4% 10% 19%
MAJOR FETAL ABNORMALITIES
1.6% 2.5% 10% 19% 24% 46%
ALIVE AND WELL
97% 93% 70% 50% 30% 15%
Note: The row “<95th centile” describes the baseline population risks. Source: From Souka et al. (2005).
Prenatal Diagnostic Procedures •
419
amniotic fluid cells obtained by amniocentesis at about 16 weeks of pregnancy. A number of other approaches to PND have since been developed, ranging from preimplantation genetic diagnosis (following in vitro fertilization), through chorion villus sampling (CVS), to fetal blood sampling, and some more experimental procedures. Naturally, parents-to-be are anxious to have results as early as possible. A desire for an early result needs to be balanced against a number of considerations which can include complexity of the procedure, both clinically and in the laboratory, procedural trauma and risks, reliability of results, cost, and the prior risk for a fetal abnormality. A useful source for the layperson is Prenatal Testing: Making Choices in Pregnancy (De Crespigny et al., 1998).
Molecular Methodologies Four particular analytical procedures have enabled a much faster return of results for common aneuploidies: FISH, QF-PCR, MLPA, and higher resolution detection of any chromosome abnormality by microarray analysis (and see also Chapter 2). FLU OR ES CEN CE IN SI T U HY BRI DI ZAT I ON
Fluorescence in situ hybridization (FISH) using multiple colored probes and targeting the chromosomes most prone to survivable aneuploidy (13, 18, 21, X, and Y) bypasses the need for culture, whether the cells are from amniotic fluid, CVS, or fetal blood, and the result can be given within the space of one working day (Klinger et al., 1992; Morris et al., 1999; Weremowicz et al., 2001; Lim et al., 2002; Baart et al., 2004). This approach is particularly useful where the need for a rapid result is more pressing, such as when fetal anomalies have been discovered at ultrasound. In one small series with particular reference to the third trimester, AviramGoldring et al. (1999) showed an aneuploidy in 23% of pregnancies with intrauterine growth retardation and structural abnormalities: five with trisomy 21, and two with trisomy 18. Feldman et al. (2000) similarly applied amniotic fluid cell FISH to high-risk pregnancies (that is, with ultrasonographic abnormalities). They detected fourteen cases of trisomy 21, ten of trisomy 18, three of trisomy 13, four of monosomy X, and one triploid, in 4193 samples over the period 1996–1998, for a total abnormality rate of 11%. A limitation is that only those specific aneuploidies being tested for (the major autosomal 420
•
P R E N ATA L D I A G N O S I S
trisomies and sex chromosome aneuploidies) are detected, and it is fair to say that conventional karyotyping is not about to be replaced by FISH (Morris et al., 1999). Waters and Waters (1999) comment that, with shortening times for standard karyotyping, there may actually be less pressure for a rapid result. While there is an attraction of being able to offer a next-day result, albeit a preliminary one, it is nevertheless worth noting that a benefit in terms of reducing anxiety and stress is only short term, and otherwise there appears to be little advantage (Boormans et al., 2010). The question arises of false-negative results. Weremowicz et al. (2001) reviewed their experience over 1992–2000, during which time they applied FISH (using probes for 13, 18, 21, X and Y) to some 8% of the 11,000 amniocentesis samples coming into their laboratory for routine karyotyping, this 8% including cases with an increased risk (abnormalities on ultrasound, serum screen result). In the whole material, of 89 potentially detectable abnormalities, 75 (84%) were found. The missed cases included 8 with an inconclusive result, one with no result, and—more importantly—5 false-negatives. Of these latter 5, the true karyotypes were trisomy 18 (2 cases) and trisomy 21 (3 cases). Technical problems related to poor hybridization efficiency (low copy number of the DNA repeats being probed, for example), and maternal blood contamination of the fluid sample, are plausible explanations. As have others, Weremowicz et al. note the usefulness of the FISH approach in being able to provide a rapid answer particularly when there are grounds for suspecting an abnormality, or if the pregnancy is more advanced, but they also emphasize the need for careful counseling so that patients are aware of the limitations. With respect to trisomy 21, Witters et al. (2002) had an encouraging record: in a similar study comprising 5049 amniotic fluid samples, in which interphase FISH was applied in parallel with conventional karyotyping, all 70 cases of trisomy 21 were detected, and no false-positive result arose. One false-positive is on record, however, probably due to technical aspects of probe hybridization (George et al., 2003). On the question of mosaicism, Van Opstal et al. (2001) note that FISH on uncultured cells may provide a more accurate picture than on cultured cells, the latter possibly being subject to selective pressure in vitro and the abnormal cells more prone to fail in culture. On the other hand, the class of amniocyte that grows preferentially in culture (namely, amniotic mesoderm)
might, according to the reinterpretation of Robinson et al. (2002), more closely reflect the true embryonic state. Focused FISH can be applied in specific circumstances. The ultrasound discovery of a cardiac outflow tract abnormality would, for example, point to the need for 22q11 analysis. A rapid diagnosis is particularly to be desired in the setting of parental heterozygosity for a chromosome rearrangement in which there may be a high risk for abnormality, and FISH can provide this. Thus, Cotter and Musci (2001) used subtelomeric probes for 5pter, 5qter, and 14qter to enable rapid diagnosis for a pregnant woman with the karyotype 46,XX,t(5;14)(p14.2; p13), she having had a previous child with cri du chat syndrome. Similarly, Pettenati et al. (2002) applied this approach in the setting of parental heterozygosity for a number of reciprocal and Robertsonian translocations. FISH tests only that segment of the chromosome to which the probe binds. Inferentially, the complete chromosome is present; but this is not necessarily so. In the case of chromosome 18, it is the centromere which the FISH probe recognizes. We have seen a case in which amniocentesis was done on the basis of a maternal age of 40 (albeit that the Down syndrome risk on second-trimester maternal serum screening had been reduced to 1 in 164), and the figure for trisomy 18 was very low (1/8030). FISH showed three chromosome 18 signals. Fetal growth and morphology on ultrasonography were normal. The couple considered whether they might request termination but wanted to await the result of karyotyping. This showed a supernumerary minute marker—barely a speck—which appeared to comprise only 18 centromere: the karyotype was 47,XX, +mar.ish der(18) (D18Z1+)de novo[13]/46,XX[4] (case of M. Pertile). The pregnancy continued. The child subsequently born was 3 years old at most recent contact, and while she manifested a familial shortness, her cognitive and personality development were entirely normal (S. Fawcett, personal communication, 2010). QUANTITATIV E F LUORESCE NCE POLYMERASE CHAIN REACTION
Quantitative fluorescence polymerase chain reaction (QF-PCR) relies on the use of molecular markers that display a high level of heterozygosity, such that the presence of three alleles—that is, a trisomy— can reliably be detected. This DNA-based test may enable rapid diagnosis. Pertl et al. (1999) performed
the preliminary studies using QF-PCR with chorionic villus samples as the test material. The sequences assessed are chromosome-specific polymorphic small tandem repeats. There being three allelic forms of a particular marker allows the assumption of trisomy for that chromosome. There may need to be some choice of markers according to population: for example, Putzova et al. (2008) have determined a robust set of markers on chromosomes 13, 18, 21, and also for the X and Y chromosomes that are most informative in a population of Czech ancestry. In the experience of their laboratory, they could report a result within one working day, for 99.5% of samples (amniocytes or chorionic villi). This methodology detects only these major aneuploidies. From a large prospective study (4,176 amniocenteses), Speevak et al. (2011) drew the conclusion that QF-PCR may be suitable for pregnancies judged at risk only for a common aneuploidy, but where the risk is deemed to be higher, G-banding would be needed to detect the small residuum of significant abnormalities missed by QF-PCR. M ULT I PL EX L I G AT I O N - D EPEN D EN T PRO B E A N A LYS I S
Multiplex ligation-dependent probe analysis (MLPA) is a PCR-based assay that can combine probes to many chromosomal loci (Schouten et al., 2002). Its main application has been in the rapid detection of aneuploidy in prenatal testing (Shaffer and Bui, 2007). In a comparison with standard chromosome analysis of 1000 amniotic fluid specimens, MLPA had 100% sensitivity and specificity for identifying common aneuploidies (Kooper et al., 2008). A targeted application enables the diagnosis of microdeletion syndromes that would otherwise have escaped detection (Konialis et al., 2011). M I CR OA R R AY A N A LYS I S
Microarray analysis either targeting specific regions (e.g., known microdeletion syndromes, subtelomeric regions, theoretical malformation pathways), or across the whole genome, has the potential to offer the widest prenatal diagnostic capacity, and usually within a few days turnaround time (South and Lamb, 2009). Retrospective studies have been helpful in demonstrating that microarray analysis will identify chromosome imbalances in prenatal samples with previously known abnormal karyotypes, or in samples from pregnancies with multiple fetal anomalies Prenatal Diagnostic Procedures •
421
(Le Caignec et al., 2005; Rickman et al., 2006). Prospective studies demonstrate the additional benefit in the detection of imbalances either missed with banding, or at a resolution too small to be seen through the light microscope (Sahoo et al., 2006, Shaffer et al., 2008; Coppinger et al., 2009a; Van den Veyver et al., 2009).
PRENATAL DIAGNOSTIC CLINICAL PROCEDURES Blastomere and Polar Body Biopsy The techniques of preimplantation genetic diagnosis have advanced considerably in recent years, and a separate chapter (Chapter 26) is devoted to a treatment of this category of “prenatal” diagnosis.
Chorionic Villus Sampling Chorionic villus sampling (CVS) is typically a firsttrimester procedure, the usual time being at 10–11 weeks gestation. (The expression “placental biopsy” could also be applied, although in practice this term is used when the testing is done in later pregnancy; see below). The earlier period of diagnosis permitted by CVS may be seen as more useful in the setting of a higher genetic risk. If a genetic abnormality is identified, and abortion is chosen, this can be, prior to 14 weeks, a more private matter, and the termination procedure is an operative intervention (curettage or suction evacuation of the uterus). Couples are more likely to make a choice for abortion when the diagnosis has been made in the first trimester (Verp et al., 1988). There is potential in CVS for diagnostic difficulty due to the occasional detection of confined placental mosaicism (which may, for some chromosomes, carry a risk also for uniparental disomy). Nonmosaic results for the common aneuploidies are, however, highly reliable (Smith et al., 1999). In experienced hands there is a high degree of safety (Brambati et al., 2002; Brun et al., 2003), and indeed in one study in which lethal fetal diagnoses were accounted for, the loss rate was only 0.23% (1/436) (Nanal et al., 2003). However, there is some preliminary evidence that, at least for nulliparous women, the procedure may carry an increased risk for preeclampsia (Grobman et al., 2009). Direct, Short-Term, and Long-Term Chorionic Villus Sampling. Chorionic villi can be analyzed directly (same day), after short-term culture (next 422
•
P R E N ATA L D I A G N O S I S
day or two), or after long-term (a week or so) culture; but most laboratories now offer only long-term CVS. Trophoblast is the source of the cell population studied at direct and short-term CVS culture. These cells are no longer extant (if they have not already been removed by trypsinization at sample receipt) after the first few days, and it is the mesenchymal core of the villus that provides the cells that are analyzed at long-term culture (and see Color Fig. 27–1). In the early 1990s there were disconcerting reports of an increased incidence of transverse limb deficiencies and tongue and jaw defects— “oromandibular-limb hypogenesis”—following early CVS (before 10 weeks, and especially up to 8 weeks). The association appeared likely to be causal, and one line of circumstantial evidence was that the rate of anomalies fell with increasing gestational stage from 9 to 11 weeks (Firth, 1997). Various mechanisms were proposed: oligohydramnios, bradycardia, hypovolemia, thromboembolism, vasoconstriction, antibody-mediated reaction, and increased apoptosis following disruption of end arteries (Luijsterburg et al., 1997). Given these observations, it became normal practice that CVS is not done earlier than 10 weeks.
Standard Amniocentesis Transabdominal amniocentesis, at about 15–17 weeks gestation, with culture of cells for chromosome analysis, has been the standard cytogenetic prenatal diagnostic procedure for over a quarter of a century. It has a high degree of safety to both mother and fetus: maternal complications, or fetal injury due to direct trauma, are practically unknown. The risk for maternal Rhesus immunization (Rh-negative mother, Rh-positive fetus) can be circumvented by administering an antibody injection. The only significant complication is a procedure-related fetal loss rate of about 0.5%. The cytogenetic results are highly reliable. The biological sources of error are, first, that maternal rather than fetal cells, or a mixture of both, are sampled. In practical terms, this rarely causes a problem. Second, fetal mosaicism may go undetected, since only a limited number of cells can feasibly be examined. Very few examples of this error are recorded. Amniotic fluid culture has a high success rate. Persutte and Lenke (1995) suggested that if amniotic cells fail to grow, for no obvious reason, there may
be a substantial risk for fetal aneuploidy (13% of 32 cases in their preliminary study). This assessment was supported in a large systematic study from London (Reid et al., 1996), in which 42 failures (1%) among 4134 amniocenteses were followed up. Complete information could be obtained on all but one of these 42 cases. Karyotyping was ultimately done in most (78%) of these failed cases, and of these 19% revealed an abnormality (comparing with a 4% abnormality rate in the whole material). The clear lesson from these studies is that women having had a failed amniocentesis culture should be offered careful review and retesting. The obvious disadvantage of standard amniocentesis with cell culture is that the results are not available until about 16–18 weeks. If the reason for the amniocentesis had been an “increased-risk” result from maternal serum screening, the procedure may not be done until 17–18 weeks, aggravating this difficulty. If a result cannot be available by 20 weeks, another procedure (placental biopsy, interphase FISH) may be worth considering. The outlook for the long-term health of the child does not differ between CVS and amniocentesis (Schaap et al., 2002). E ARLY AMN IO CEN TESI S
In the late 1980s early (10–13 weeks) amniocentesis was proposed as an alternative to CVS. In a carefully controlled comparison, Nicolaides et al. (1994) found a 2%–3% additional fetal loss rate in early amniocentesis and, possibly, a higher incidence of talipes among subsequently born children. Daniel et al. (1998), comparing 10–14 week procedures with 15 weeks and upward, observed that the early amniocentesis samples were not quite as satisfactory, multiple needle insertions were more often required, and the pregnancy loss rate was greater. On the whole, the differences were not great, other than the loss rate of 2.2% in the early group compared with only 0.6% in the midtrimester group. Similar figures are reported in Collins et al. (1998). In the Canadian Early and Midtrimester Amniocentesis Trial, the findings for 11wk 0d through to 13wk 6d were somewhat more disconcerting, with more complications, and a higher culture failure rate (Delisle and Wilson, 1999). The procedure is rarely undertaken now. FE TAL B LO O D SA MPLI NG
Fetal blood is aspirated by direct puncture of a blood vessel, usually in the umbilical cord (cordocentesis).
Before FISH analysis of uncultured cells (see above) came to be more widely used, cordocentesis was useful when speed of diagnosis was of the essence, in the setting of the detection of a fetal anomaly at ~18-week ultrasonography. The procedure once had a role in assisting resolution of mosaicism in amniotic fluid culture (Shalev et al., 1994), but this has largely been replaced by the use of FISH. PLACENTAL BIOPSY
In principle, this is the same as first-trimester CVS. The placenta is sampled by a transabdominal approach, and this is a straightforward procedure. The main application had been when a rapid result was needed, although newer methodologies (see above) have now largely by-passed that imperative. An insufficient amount of amniotic fluid remains an indication.
Experimental Approaches, Including “Noninvasive Prenatal Diagnosis” A variety of experimental approaches to clinical and laboratory prenatal diagnosis have been, or continue to be, under trial, and for the sake of completeness we mention them here. No procedure is likely to replace amniocentesis or CVS, at least in the near future. But the counselor will certainly want to keep aware of developments in the noninvasive, or at least less invasive, prenatal diagnostic tools. Being able to separate out fetal DNA, after something as simple as the taking of a maternal blood sample, has an obvious attraction. A Dutch group have already tested consumer views, and they find considerable support, in principle, from a population of pregnant women, although somewhat less so from a comparison cohort of female masters students (Kooij et al., 2009). As for sampling via the cervical canal, women may differ in their views as to the degree of invasiveness due to the insertion of an instrument; but the precedent of the Pap smear, and in contrast to an amniocentesis or CVS needle, might color acceptance of this approach. F E TA L CEL L A N D F R EE F ETA L D N A I SO L AT I O N F R O M M AT ER N A L B LO O D
The two important cell types that are released from the fetal tissue into the maternal circulation are the nucleated red blood cell and the trophoblast, and the latter cell releases fetal DNA into the maternal Prenatal Diagnostic Procedures •
423
bloodstream (Dhallan et al., 2007; Maron and Bianchi, 2007). Research into the use of these materials for prenatal diagnosis is monitored by the National Institutes of Health Fetal Cell Study (NIFTY) group in the United States and the Special Advances in Fetal Evaluation (SAFE) group in Europe (Bianchi and Hanson, 2006). Various sophisticated means are employed to separate out these components from a sample of maternal blood. DNA has shown more promise than cells as the diagnostic material (Chiu et al., 2009; Lun et al., 2011). In a large study based upon 753 pregnancies at increased risk for trisomy 21, Chiu et al. (2011) applied the methodology of massively parallel genomic (“nextgeneration”) sequencing. According to a chosen protocol applied to subset of these, they were able to identify all (86/86) affected pregnancies (confirmed on classical karyotyping), but with 2% of 146 non– trisomy 21 pregnancies misidentified as being affected. Celocentesis. The extra-embryonic celom, which exists during the first trimester, is a source for (nondividing) cells originating from extra-embryonic mesoderm. Given its anatomical continuity with the cytotrophoblast (Color Fig. 27–1), Makrydimas et al. (2006) comment that it could be conceived of as “a liquid extension of the placenta.” The procedure has the attraction of an earlier timing (6–9 weeks) than CVS, but there is a high postprocedure miscarriage rate (Ross et al., 1997). Chatzimeletiou et al. (2005) have assessed its technical feasibility using FISH, prior to elective termination in a series of 12 pregnancies, interrogating chromosomes 3, 7, 9, 13, 16, 17, 18, 21, 22, X and Y, and the result was to hand within a few hours. Jouannic et al. (2008), in a similar series, noted the problems of low (or zero) cell numbers in the sample, and a risk of maternal cell contamination; they speculate that microarray analysis might improve the diagnostic capacity. Cystic Hygroma and Pleural Effusion Fluid. Cystic hygroma has a strong association with fetal aneuploidy, especially monosomy X. A concomitant oligohydramnios may make amniocentesis difficult. Fluid from cystic hygroma and pleural effusion contains lymphocytes and these cells can be cytogenetically analyzed within the timeframe of a few days. In one small series, 3 out of 4 cystic hygroma analyses showed aneuploidy (trisomy 21, monosomy X) (Costa et al., 1995).
424
•
P R E N ATA L D I A G N O S I S
Cervical Lavage or Cytobrush. Trophoblast cells may migrate from the confines of the uterine cavity and enter the endocervical canal, and they can be collected for molecular analysis by endocervical irrigation and aspiration (lavage), or by insertion of a “cytobrush” (Bischoff and Simpson, 2006). The attraction, in principle, is of early (7 weeks) diagnosis and a (relatively) noninvasive procedure. Cells of fetal origin are greatly outnumbered by maternal cells, and cleanly isolating the few trophoblastic cells is a difficult challenge. One way around this problem is simply to analyze all the cells retrieved (by X, Y, and 21 FISH) and to assume that any trisomy detected would be fetal (Sifakis et al., 2010); the question of a false-negative XX result from such an approach would remain open. Proteomic Fingerprinting. Proteomic fingerprinting of amniotic fluid assesses the expression profile of proteins coded from specific chromosomes, or otherwise expressed in the context of a specific aneuploidy, and this could be considered as a functional assay for trisomy (Mange et al., 2008; Koster et al., 2010).
“Primum non nocere” “First, do no harm” is a cornerstone of medical practice. Yet, almost inevitably, having a prenatal diagnostic procedure causes anxiety. Rothman (1988), in her book The Tentative Pregnancy, is particularly critical of what she sees as a medicalized distortion of the normal process of being pregnant. Hodge (1989) describes her personal experience of Waiting for the Amniocentesis, and we reproduce her letter in full: I drafted the following letter to the editor one week before I expected to hear the results of my amniocentesis: “I am 40 years old and 19 weeks pregnant with what will presumably be my third child. I am on the basic science faculty of a medical school. When I teach medical students about amniocentesis, I occasionally mention the difficulty for the woman of having to wait until well into the second trimester to receive her results. “I am in that situation myself now, awaiting my results. And before experiencing it, I was unprepared for two phenomena. One was
just how difficult the wait is. Pregnancy is always a time of waiting, but now time has slowed down to an extent I did not anticipate. The other, more disturbing phenomenon is how the waiting has affected my attitude toward the pregnancy. At many levels I deny that I really am pregnant ‘until after we get the results.’ I ignore the flutterings and kicks I feel; I talk of ‘if ’ rather than ‘when’ the baby comes; I am reluctant to admit to others that I am pregnant. I dream frequently and grimly about second-trimester abortions. In some sense I am holding back on ‘bonding’ with this child-to-be. This represents an unanticipated negative side effect of diagnostic amniocentesis. And all this, even though my risk of carrying a chromosomal abnormality is less than 2 percent. “I presume I am not alone in these reactions, yet I have not seen this problem mentioned in the literature, nor did my physician or genetic counselor discuss it with me. I am writing now to bring it to the attention of clinicians with pregnant patients undergoing diagnostic amniocentesis. I suggest to both clinicians and their patients that, when weighing the relative risk and benefits of prenatal diagnosis performed later (amniocentesis) as compared with earlier (chorionic villus biopsy), they not underestimate the negative effects of a 4½ month wait before the woman knows if she is ‘really’ pregnant.” The next day, before I had mailed this letter, I received the results, and unfortunately they were the dreaded ones: trisomy 21. I have since then had the grim second-trimester abortion. From my current perspective of grief and shock, I encourage clinicians to help their patients avoid the denial described in my letter. My husband and I spared ourselves no pain by holding back emotionally. It has become a cultural expectation that one will keep one’s pregnancy a secret until one has had the “all clear” from the amnio. One reasons, “If we get a bad result, we won’t have to tell anyone.” But I now believe that reasoning is wrong. After our bad result, my husband
and I did tell everyone. Sympathy and support from our friends, family, and colleagues have helped us to survive the ordeal of aborting a wanted pregnancy. By keeping the loss a secret, we would have cut ourselves off from such support when the feared outcome did happen. Not every couple will react this way, some preferring to keep their personal affairs private, but many will. The counselor needs to acknowledge these criticisms, and to rise to the challenge of providing a sympathetic and skilful service to clients/patients, according to their varying responses to deciding to have, to undergoing, and to waiting for the results of prenatal diagnosis, and then supporting those who do get an abnormal result. These issues are addressed in detail in Prenatal Diagnosis: The Human Side (Abramsky and Chapple, 2004). A considerable fraction of pregnant women are, in any event, opposed to invasive prenatal testing. In a study of pregnant women (age 37 and older) who had not undergone prenatal diagnosis in Victoria, Australia, 33% had actively declined, with the two main reasons being concern about the safety of the test, and a conviction that they would not in any event have a termination. Another 6% had never been offered testing, these being for the most part women from minority groups, with single women also overrepresented, and it is a challenge to see that all who might wish to have the choice of prenatal diagnosis are indeed given it (Halliday et al., 2001). A practical question is pain: the thought of insertion of a needle, or of a catheter, sufficiently deeply to sample a pregnancy, would naturally be cause for some apprehension. Csaba et al. (2006) surveyed a number of women undergoing prenatal diagnosis in New York, asking them to quantify their anxiety ahead of the procedure (transabdominal CVS, transcervical CVS, or amniocentesis), and their perception of pain immediately afterward. In each procedure, the pain was typically seen as “mild,” and threequarters of the women thought it was the same or less painful than they had been expecting. Those who were more anxious—mostly the younger and nulliparous—felt the pain more keenly, and thus special reassurance should be given to this group.
Prenatal Diagnostic Procedures •
425
26 preimplantation genetic diagnosis
CHROMOSOMAL PREIMPLANTATION genetic diagnosis is done in the setting of in vitro fertilization, and in principle it enables an unaffected embryo to be transferred to the uterus, some 3 to 5 days post fertilization. Thus, for couples facing a high genetic risk, the risk can be bypassed; and the prospect of pregnancy termination for the reason of genetic abnormality can be avoided. Advances in the late twentieth century in the fields of in vitro fertilization (IVF), human embryo culture and manipulation, molecular genetics, and fluorescence in situ hybridization (FISH), set the stage for the development of preimplantation genetic diagnosis (PGD). From an essentially researchbased exercise in a very few laboratories in the early 1990s, it has progressed to being, in the 2010s, a diagnostic tool available through a number of larger IVF clinics. PGD is applied in two main genetic settings: for the diagnosis of chromosome disorders, and for the detection of a mendelian condition.
Initially, the two categories were distinguished by the methodology applied: FISH in the former, DNA testing in the latter; a distinction that is now blurring, as molecular methodologies advance. There has arisen a praiseworthy tradition of excellent communication between the major centers that do this work, with the majority contributing their data to an international clearing house (under the aegis of ESHRE, the European Society for Human Reproduction and Embryology), and detailed analyses of the accumulated experience of the participating clinics are documented in the annual reports of the ESHRE PGD consortium, which appear in the journal Human Reproduction (Harper et al., 2010b); and the International Society for Prenatal Diagnosis sponsors a regular conference devoted to PGD. Thus, new knowledge from the leading centers can translate readily into improved services to patients worldwide. Chromosomal PGD is typically done on a blastomere (single cell) from a 6–10 cell embryo1 at day 3.
1 We mentioned the power of language in referring to babies or fetuses, and mothers and pregnant women, on p. 418. Embryo is another word with a laden identity, and expressions such as “embryo destruction” can carry emotional weight. In fact, the embryo proper
426
•
Alternative approaches are blastocyst biopsy at day 5 and polar body biopsy. The selected embryo is then transferred that same day (day 3 transfer) or maintained in culture a little longer (day 5 or 6 transfer). In principle, and barring the presence of mosaicism (a very important issue; see later), the pregnancy can proceed in the knowledge that the baby will be unaffected. There are two main categories of chromosomal PGD: focused PGD with respect to a particular parental translocation, or other rearrangement; and PGD being done as a general aneuploidy screen (PGD-AS).
PATIENTS AND CIRCUMSTANCES WHERE CHROMOSOMAL PREIMPLANTATION GENETIC DIAGNOSIS MAY BE APPROPRIATE Carriers of Balanced Rearrangements A parent who is the carrier of a balanced rearrangement typically has a high risk to produce unbalanced embryos, as discussed at length in earlier chapters. Particularly in the context of an unfortunate reproductive history, often with several miscarriages, or with one or more terminations following conventional prenatal diagnosis of an unbalanced fetal karyotype, the attraction of PGD is obvious: only an embryo with a normal or balanced chromosomal constitution is transferred. The two main categories are reciprocal (rcp) and Robertsonian (rob) translocations. In principle, the outcomes should be improved; and in practice, this is the case (Munné, 2005; Keymolen et al., 2009). An impressive report comes from Otani et al. (2006), who assessed PGD in 33 couples having had several miscarriages and no liveborn children, from a total of 117 pregnancies, and one of the couple being a translocation heterozygote. Thus, in their prior reproductive history (typically over several years), there had been a 100% pregnancy loss. Following PGD (an average of 1.24 cycles per patient), a total of 20/88 embryos from rob carriers, and 86/491 from rcp carriers, were diagnosed as normal/balanced, these comprising
only 18% of the total (again attesting to the high genetic risk). Of the 19 pregnancies subsequently resulting from transfer of normal/balanced embryos, just one (5%) miscarried; the other 18 pregnancies had either proceeded into the second trimester or had culminated in live birth. 100% loss versus 5% is a notable contrast. This very considerable improvement does imply that many of these couples would otherwise have had no impediment to fertility (although not all couples had been able, at the time of the study, to achieve a pregnancy: as applies, of course, to all IVF). A high rate of unbalanced gametes would logically translate into a similarly high risk for unbalanced embryos. A stringent assessment of a link between the proportion of unbalanced sperm and the outcome at PGD comes from Escudero et al. (2003). They noted that a fraction of greater than 60% unbalanced sperm was associated with a poor reproductive prognosis, and that biopsied blastomeres had a somewhat higher abnormality rate than did the sperm; and they devised a formula to estimate the relationship between these rates. The formula is A = – 55 + (1.9 × B), where A is the percentage of abnormal embryos, and B the percentage of abnormal sperm. We may apply this formula to the example in Wiland et al. (2008), concerning their study of a man heterozygous for t(2;7)(p11.2;q22), he and his partner (and other family members) having suffered recurrent miscarriage. Of his sperm, 66% revealed malsegregation; whence A = – 55 + (1.9 × 66) = 70%. The actual observation at PGD was 57% abnormal embryos, but this was based on the small numbers of 4 out of 7 embryos being unbalanced.2 Had the rate been 5 of 7, the fraction would have been 71%. But in practice, sperm analysis is not routinely undertaken; the essential thing is PGD result.
Women of Older Childbearing Age and Preimplantation Genetic Diagnosis–Aneuploidy Screening Given the maternal age effect in chromosomal abnormality, it had initially been supposed, and quite
has yet to develop—this only happens once the primitive streak has started to differentiate. Jones and Veeck (2002) debate these fine points and propose that “pre-embryo” is a more accurate term for the period from conception through to the appearance of the primitive streak, that is, from 0 to 14 days. Another word with credible currency for the stage at about day 4 is morula. On about day 5, the next stage is the blastocyst (and see Fig. 27–1, 1 and 2), and this term is specifically applied to biopsy undertaken at this time. 2 In this case, two of the three normal/balanced embryos were transferred, with a triplet pregnancy resulting. Two of the fetuses were monozygous heterozygous male twins; for obstetric reasons, these two fetuses were selectively terminated, and the remaining 46,XX fetus carried to term, and a normal girl born.
Preimplantation Genetic Diagnosis •
427
reasonably so, that PGD for aneuploidy screening (PGD-AS) would be valuable for older mothers; more often in the setting of difficulty in conceiving, or with a history of miscarriage. In other words, older mothers who might otherwise have been considering IVF would have the option of adding PGD-AS to the procedure; and eliminating embryos with a trisomy would “surely” improve their chances of a chromosomally normal baby (Munné et al., 2002a). Apparent Poor Results from PGD-AS. But the early promise of PGD-AS faded as more units began to offer this service. One large and stringent trial (multicenter, randomized, double-blind controlled), conducted over 2003–2007, comparing PGD with standard IVF in 408 women, of age range 35-41, showed a clear lessening in success (a 25% vs. 37% pregnancy rate), in those receiving PGD-AS (Mastenbroek et al., 2007). Twisk et al. (2008) reached a similar conclusion against a favorable effect due to PGDAS. Similarly, PGD-AS from blastocyst biopsy may have less success than simple blastocyst transfer ( Jansen et al., 2008). This paradox demanded explanation (Donoso et al., 2007). Overinterpreted Mosaicism? Vanneste et al. (2009a,b) accounted for these observations as follows. The rate of mitotic error during the first 3 days of postfertilization existence is so high—much higher than that due to meiotic error—that biopsy of a single blastomere may fail to give a true picture of the karyotype of the conceptus. Most of the cells may be abnormal: “chaotic mosaicism” (and see later). The abnormalities comprise both simple aneuploidies and deletions/duplications due to the effects of a “breakage-fusion-bridge cycle.” But the small minority of normal cells may be the ones that endure, and which, following implantation, are the ones that give rise to the embryo, with the abnormal cells cast aside (Barbash-Hazan et al., 2009). According to this view, these first three waves of mitosis, that give 2, 4, and then 8 cells, may be a particularly vulnerable stage in human development; but also a stage during which, if a single biopsied cell is shown to be abnormal, the risk can be overinterpreted, and the interpretation misleading. Aneuploidy, in mosaic state, may be rather common, and many embryos might be, by the criterion of PGD-AS, rejected: and yet some could have produced a “take-home baby” (in the jargon of fertility clinics). Some support for this interpretation might come from Hanson et al. (2009), who examined the accuracy of the chromosomal diagnosis at PGD-AS, by 428
•
P R E N ATA L D I A G N O S I S
analyzing the constitutions of 173 embryos that had tested aneuploid at blastomere biopsy. Only 10% (the authors’ comment that this correlation was “rather low” somewhat of an understatement) proved to have the exact same diagnosis that had been made at PGD. One-third did, however, have the same abnormality as at PGD, along with other abnormalities; and almost all (96%) of the embryos did have a chromosomal abnormality of some sort, albeit in many, different from the PGD diagnosis. From these data, the conclusion might be drawn that PGD-AS, as a diagnostic procedure, has been thwarted by the biology of chaotic mosaicism. Ascertainment Bias Due to Only Discarded Embryos Being Analyzed? Wilton et al. (2009) emphasize the very high diagnostic accuracy when the test endpoint is not the analysis of an untransferred embryo, but the baby. Of course, it is typically the “goodlooking” embryos that are chosen for transfer, and the discarded ones whose study reveals the chaotic mosaicism. Perhaps good-looking embryos, if we could but observe their tubal journey in vivo, might, in contradistinction to the interpretation outlined earlier, quite often present a picture of nonmosaic chromosomal normality. In one review in which a number of PGD-normal embryos had not been transferred, none of these turned out to be abnormal (Staessen et al., 2004). And Moayeri et al. (2008) observed that the appearance of the embryo, according to the “day-3 morphology score,” may, in an older mother, predict an improved chance for euploidy at PGD-AS. Confounding Due to FISH Artifact? Treff et al. (2010a) propose that mosaicism at PGD may be more apparent than real: simply for technical reasons inherent in the methodology, FISH may be presenting a false picture (and see section on “The Technical Challenge of FISH” that follows). If the same embryos are tested in parallel with FISH and SNP-microarray, mosaicism in the latter group falls to much lower levels, in a range of 25%–30%. Diagnosis of aneuploidy by FISH may be disturbingly inaccurate: for example, in a separate study of day 3 embryos called as trisomic by FISH, nearly half were actually euploid, when four different sections of the day 5 blastocyst (three from the trophoblast, one from the inner cell mass) were examined by microarray (Northrup et al., 2010). The normality of the trophoblast in all sections undermined the theory that aneuploid cells might be sequestered to potential placental tissue. Treff et al. (2010a) suggest that “it is becoming clear that FISH-based technology is
inadequate for the diagnosis of aneuploidy in human embryos.” The Need for Clinical Trials. Be these observations and conclusions as they may, given the disappointing results in practice from PGD-AS, an expert panel under the aegis of ESHRE (Harper et al., 2010a) has concluded that “the most effective way to resolve the debate about the usefulness of PGS is to perform well-designed and well-executed randomized clinical trials.” Such trials, they recommend, should distinguish time of biopsy (cleavage stage, polar body, or blastocyst), the methodology applied (FISH versus microarray), and appropriate end-point (delivery rates). The PGD community will await the findings of such a trial with much interest. The early reports coming from SNP-array aneuploidy screening do offer grounds for optimism, and Treff et al. (2010b) claim an impressive 98.6% accuracy of diagnosis.
Recurrent Miscarriage and Preimplantation Genetic Diagnosis–Aneuploidy Screening Some couples who are chromosomally normal, but who have had a history of multiple miscarriage, may benefit from PGD-AS (Rubio et al., 2003). The fraction of spontaneous abortion that is due to chromosomal abnormality has been revised upwards in recent years (Menasha et al., 2005, and see p. 384); and some miscarrying couples may have been having recurring aneuploidies. Ferraretti et al. (2004) showed in a series of PGD patients presenting for repeated cycles of treatment, for the reasons of either older age (>37 years), or of past implantation failure, that previous euploidy or aneuploidy predicted future euploidy or aneuploidy. Similarly, Munné et al. (2004), demonstrated that, at least for younger (<35 years) women, a previous trisomic pregnancy predicted an increased risk for future aneuploidy. The women in this study, coming forward for PGD-AS, had only 29% of embryos euploid, on a limited range of chromosomes tested (X, Y, 13, 18, and 21, and in most cases also 15, 16, 17, and 22), compared with 52% in those having the indication of gender selection due to an X-linked condition (and whose fertility would presumably have been unimpaired). Increasing the FISH panel would enable more aneuploid embryos to be recognized as such (Lathi et al., 2008), or with microarray, the whole karyotype is examinable. While the interpretation may be a little clouded, considering that there is a presumed meiotic basis in
a previous aneuploid pregnancy, but with the possibility of a mitotic generation of a PGD aneuploidy(ies), nevertheless, the inference that PGD may have value in the setting of a history of aneuploid miscarriage is not unreasonable. The ESHRE panel mentioned earlier has taken a conservative stance and proposes that a true view of the appropriateness of PGD-AS in the setting of recurrent miscarriage requires the insight that only a randomized controlled trial would provide (Harper et al., 2010a). Gender Selection. Gender diagnosis at PGD, with the use of X and Y FISH probes, may be appropriate in the context of a sex-related genetic risk, whether mendelian or nonmendelian, an example of the latter being autism (Amor and Cameron, 2008).
CLINICAL AND LABORATORY PROCEDURES From the earlier categories of patient, two major karyotypic classes of couple are to be considered: couples one of whom carries a balanced chromosomal rearrangement; and those who have normal chromosomes, and who may have chosen “aneuploidy screening”. The former group will command most of our attention, although of course they are the smaller group. Those who make the decision to embark upon chromosomal PGD—and for whom the laboratory have advised that testing, in their particular case, would be feasible—will need to enroll (if not already) in an IVF program. If FISH is to be the methodology, close liaison with the laboratory is required, in order that the scientist can have the appropriate probes prepared and ready for use on the day. The timeframe for having hyperstimulation of ovulation is established from the woman’s menstrual cycle, and ovum “pickup” is conducted by transvaginal endoscopy under ultrasound guidance. Given the high risk of abnormal malsegregants typically associated with chromosome rearrangements, it may be useful in this situation to employ a stimulation protocol designed to maximize the numbers of ova produced (Fridström et al., 2001), albeit that a concern that stimulation might of itself tend toward aneuploidy needs to be weighed (Baart et al., 2007). The ova are collected, and then exposed to sperm in vitro. If the couple are of otherwise normal fertility, simple mixing with the male partner’s sperm suffices. With some forms of male infertility, and this is Preimplantation Genetic Diagnosis •
429
quite often the case in the male heterozygous for a chromosome rearrangement, ICSI3 is needed. On day 1, around 18 hours after exposure to sperm, the oöcytes are checked for the presence of two pronuclei and two polar bodies, as evidence that fertilization4 has occurred. They are then returned to tissue culture medium; in a few hours syngamy will occur, and over the next 48 hours the first few mitoses will have produced cleavage-stage embryos of 6–10 cells.
Standard Day-3 Blastomere Biopsy On day 3, in the morning, one or at most two cells (blastomeres) are removed from each embryo, under the inverted microscope.5 This requires a hole to be made in the “shell” (the zona pellucida, which has not yet been cast off ), the cells being extracted by very gentle suction. These cells are subject to FISH or microarray analysis, in order to determine whether they have a normal/balanced chromosome constitution or an unbalanced form of the rearrangement. One or two embryos shown to be chromosomally normal/balanced are then transferred to the uterus, on the afternoon of day 3 or the morning of day 4, and with luck one6 will develop into a normal infant. The remaining embryos with a normal/ balanced chromosomal complement will usually be cryopreserved, in case the first embryo transfer does not result in a pregnancy, and perhaps for a second pregnancy further in the future. The process is outlined in Figure 26–1.
abnormalities, including most monosomies and some extensive mosaics, impose a lethal burden (Clouston et al., 2002). The chances of successful transfer might, in principle, be better if the embryo has declared itself capable of developing this far, or, as Johnson et al. (2010a) put it, if it has “had the opportunity to ‘self-correct.’” There might seem some attraction in delaying PGD until the blastocyst is forming, since cell number has increased, and differentiation between inner cell mass and trophoblast has begun, allowing sampling to be focused on the trophectoderm (in a tissue-origin sense, a very early chorionic villus sampling). This involves making a hole in the zona pellucida and allowing a small part of the lining of the blastocele cavity to herniate through (“assisted hatching”), and part of this tiny bulge could be excised by laser, or teased away by manipulation (Fig. 26–2). In an array-based analysis, using embryos from a youngish cohort of couples (average maternal age = 31), Johnson et al. (2010a) examined blastocysts from which they were able to dissect out the trophectoderm and the inner cell mass. Encouragingly, almost all were concordant as to karyotype between these two tissues (and note the similar findings from Northrup et al. 2010, discussed earlier). And perhaps reflecting the younger age profile, the considerable majority (80%) of embryos were euploid. Against this approach, it is a given that the maternal genital tract offers a better environment for growth than does an IVF container, and a better chance for survival. A controversial question is whether this longer incubation in vitro might disturb the epigenetic state of the embryo.
Blastocyst Biopsy If the embryo is incubated for 1–2 more days, it advances through the morula and early blastocyst stages. Considerable selection pressure applies during this short period, and many chromosomal
Polar Body Analysis Polar body (PB) genetic analysis (satisfyingly requiring recall of some elementary facts of biology) has been used for PGD (or “preconception diagnosis”)
3 For the record, pregnancies conceived following ICSI are not more prone to confined placental mosaicism than is the general population (Minor et al., 2006.) 4 Since fertilization in vitro can be observed as it actually happens, the fine detail of the process can be appreciated. The first act is penetration of the ovum by the sperm. To the embryologist, this is only the prelude to conception; the true moment of conception is the point at which the male and female pronuclei fuse, their chromosomes aligning on a common metaphase plate (“syngamy”). Once that event has taken place, the zygote has come into existence. At the first mitosis, it loses that name, and becomes, in IVF parlance, a “cleavage-stage embryo,” or simply an embryo (or more pedantically but perhaps usefully, as commented earlier, a pre-embryo). 5 Removing two cells from an embryo with seven or more cells has been considered not to affect the potential of the embryo to develop (Van de Velde et al., 2000). 6 If two embryos are transferred, this is not designed to produce twins (for whom there is an increased obstetric risk), but rather to improve the odds that one will succeed. PGD may allow a lesser number of embryos to be transferred—ideally just one (“elective single embryo transfer,” eSET)—thus reducing the likelihood of multiple pregnancy.
430
•
P R E N ATA L D I A G N O S I S
(a)
(b)
(c)
(d)
(e)
(f)
FIGURE 26–1 The process of in vitro fertilization (IVF) (with or without intracytoplasmic sperm injection [ICSI]) and preimplantation genetic diagnosis (PGD) at the day-3 stage. (a) Oöcytes are obtained from the woman, and sperm from the man (by testicular aspiration, if necessary). (b) Oöcytes and sperm are mixed in vitro; or, single sperm are injected into an oöcyte (ICSI). (c) Syngamy, the fusion of male and female pronuclei, occurs. After incubation for 3 days, (d) one or two7 blastomeres are removed from the embryo, and (e) these cells are then subject to chromosomal analysis. (f) Normal (or balanced) embryos are chosen for transfer to the uterus, or possibly for cryopreservation for a future transfer.
7
The live-birth rate is less when two cells are removed (De Vos et al., 2009).
FIGURE 26–2 The process of blastocyst biopsy, at day 5–6. The blastocyst is held in place by the suction pipette on the left, which is applied directly to the zona pellucida (the “shell” that invests the blastocyst, “inherited” from the ovum; see Fig. 26–3). Trophectoderm (TE) has herniated through a laser-generated hole, visible in this view at 3 o’clock in the zona pellucida; the inner cell mass (ICM) remains comfortably within the zona pellucida. Suction will be applied through the biopsy pipette (right), and about five cells from the TE gently teased off. (From S. J. McArthur et al. 2008, Blastocyst trophectoderm biopsy and preimplantation genetic diagnosis for familial monogenic disorders and chromosomal translocations, Prenatal Diagnosis 28: 434–442. Courtesy R. P. S. Jansen; reproduced with the permission of Wiley-Blackwell.)
in a few research laboratories, and legal or logistic constraints against PGD in some jurisdictions have propelled interest (Landwehr et al., 2008; Montag et al., 2009). The approach has typically been FISH based. The process of biopsy is illustrated in Figure 26–3. PB analysis allows a focus on the vulnerable gamete, that is, the ovum, the great majority of meiotic nondisjunction occurring here. Disomic or nullisomic gametes could be identified, and thus excluded from fertilization. By way of example, imagine that the asterisked gametocyte in Figure 3–3a (in Chapter 3) is the first PB (PB1), and that the two chromosomes shown within it are chromosome 18s: that is, PB1 is disomic 18. The “empty” gamete to the right, therefore, would be a nullisomic 18 oöcyte, and thus of course to be discarded. A nullisomic second PB (PB2) (one of the empty cells in the next row) should provide corroboration. The reader may also determine, on study of Figure 3–6 with respect to predivision of 432
•
P R E N ATA L D I A G N O S I S
chromosomes at meiosis, why analysis of PB2 alone could in some instances mislead. The cell labeled “disomic gamete” in this figure could be the oöcyte, but PB2, represented by the cell next to it, shows a normal monosomy. Both PBs together can enable the full picture to be deduced, and the disposition of all four chromatids can be accounted for, provided probes are used that enable the distinction between single chromatids and double-chromatid chromosomes. A euploid egg might become, after fertilization, an embryo with some aneuploid cells (“chaotic mosaicism,” as elsewhere discussed in this chapter). But provided the euploid cell(s) carry on through and give rise to the inner cell mass, then this transient mosaicism will have been unknown, and unimportant. Thus, a theoretical advantage of PB diagnosis is that the uncertainty relating to aneuploidy screening at PGD, due to biopsy of a possibly unrepresentative aneuploid blastomere, could be avoided (Geraedts et al., 2010). Landwehr et al. (2008), in a research setting, have applied PB analysis in the management of couples presenting with repeated implantation failure, or for advanced maternal age, and, using comparative genomic hybridization (CGH) and thus able to interrogate every chromosome, they discovered an aneuploidy rate of 75%. This figure is very different from the corresponding figure of 22% in the series of Fragouli et al. (2006b); differences in the reproductive backgrounds and ages of the couples coming forward likely underlie this seeming incongruity (indeed, half of the couples in Fragouli et al. were attending because of male factor infertility). SEG R EG AT I O N A N A LYS I S A N D F I SH PR O B E SEL ECT I O N
Analysis of segregation patterns of structural rearrangements has been discussed extensively in earlier chapters. The specific PGD-related risks for reciprocal and Robertsonian translocations are noted in Chapter 5 and Chapter 7, these two forms accounting for the substantial majority of “chromosomal” PGD patients. When choosing FISH probes for a particular PGD, each and every possible segregation outcome, as noted later, must be considered. The pattern of FISH signals that each outcome would generate, and the certainty of being able to distinguish a balanced or normal chromosome constitution, need to be carefully thought through. For the common case of the autosomal translocation, and using the example of a translocation 46,XY,t(14;18),
FIGURE 26–3 The process of polar body biopsy. (Left) The egg is held in place by a suction pipette, which is applied directly to the zona pellucida (the “shell” that invests the egg itself). The first and second polar bodies are located in the space between the zona pellucida and the egg’s cell membrane (the latter seen in dark circular outline). The egg is manipulated so that the polar bodies are at the 1 o’clock position. (Right) The pipette has entered a laser-cut hole in the zona pellucida, and the two polar bodies have been aspirated into its lumen. (From M. Montag et al., 2009, Polar body biopsy: a viable alternative to preimplantation genetic diagnosis and screening, Reproductive Biomedicine Online, 18 Suppl 1:6–11. Courtesy M. Montag; reproduced with the permission of Elsevier.)
we set out in Figure 26–4 the full range of possible blastomere combinations. As noted earlier, the more embryos that can be biopsied, the better, since the odds for unbalanced embryos are high and may vary unpredictably from cycle to cycle (Fridström et al., 2001). Probes are designed which hybridize to judiciously chosen parts of the chromosomes of interest. With most simple reciprocal translocations three probes will, in general, be required: two that hybridize to a point within the translocated segments, and one to one of the centromeres. Pericentric inversions lead only to two unbalanced forms, and these can be accounted for by the use of a subtelomeric probe at either end of the chromosome, and a centromeric probe (Escudero et al., 2001). The most readily available commercial markers for recognition of the translocated segment are subtelomeric probes (Scriven et al., 1998). Observing the number and (to some extent) the disposition of colored spots in the nucleus of a blastomere removed from the IVF embryo allows a deduction of the chromosome complement. Probes to α-satellites enable rapid (2–3 hour) detection of the centromeres, although the single- or low-copy probes for telomeres take longer to hybridize, necessitating a late day 3 or early
day 4 transfer (Delhanty and Conn, 2001). It is necessary to show that the probes give clear and unequivocal signals on interphase nuclei and chromosomes from the carrier parent, ahead of proceeding with IVF and PGD. If there is a living individual with the imbalanced state, the opportunity should be taken to check that the probes would give a correct interpretation. These points on probe selection are dealt with more expansively in Delhanty and Conn (2001). The Technical Challenge of FISH. Single (or at most two) cell FISH requires consummate skill on the part of the cytogeneticists and embryologists who do this work. Even the most technically adept scientist, however, cannot achieve 100% of resolvable FISH signal on all chromosomal targets. This is not normally a problem for other applications where many nuclei or metaphases are available for study. In PGD it is an important, and perhaps a critical limitation. Consider the impediments to success in a “simple” PGD for a typical reciprocal translocation. Suppose three probes are to be used, each probe hybridizing to two chromosomal sites (on two normal chromosomes, or on the normal and derivative chromosomes, Preimplantation Genetic Diagnosis •
433
(a)
14
(b)
der(14)
der(18)
18
14 der(14) der(18) 18
2:2
3:1
Alt.
Tertiary mono/ trisomy
Adj.-1
Inter. mono/ trisomy
Adj.-2
Tertiary mono/ trisomy
4:0
Inter. mono/ trisomy
Carrier father
Normal mother
Blastomere
FIGURE 26–4 (a) Partial karyotype of a translocation 46,XY,t(14;18)(q24.1;q21.3) that had been the presumed basis of a series of miscarriages, leading the couple to seek preimplantation genetic diagnosis (PGD). The colored spots on the cartoon karyotype reflect those seen in the same chromosomal sites on the fluorescence in situ hybridization (FISH) analysis. (b) PGD using FISH for this translocation. The probes hybridize to the 14q telomere (red), the 18q telomere (yellow), and the 18 centromere (blue). Upper, the expected patterns of FISH probing for the 16 possible segregant outcomes (cf. Figure 5–4 in Chapter 5). Lower, FISH on parental lymphocytes shows the patterns of dots to be the same either in the carrier parent (father, left) or the noncarrier (mother, middle). FISH analysis of a biopsied blastomere (right) shows the pattern of the normal or balanced state, and thus this is a suitable embryo for transfer. The depictions of the upper panel are artificially produced; the lymphocytes and blastomere of the lower panel are actual observations. Note that in each blastomere, three dots, one of each color, are due to the gamete from the noncarrier parent, and the remaining dots represent the chromosomal contribution from the carrier parent. (Case of E. Baker.) See also separate color insert.
respectively), to give six hybridization spots in total from the normal or balanced embryo. Supposing that the hybridization efficiency of each probe is 98%, the probability of the normal or balanced embryo giving six correct signals in one cell is (0.98)6 = 0.89. These facts of simple arithmetic point to the risk that a normal or balanced embryo could be diagnosed as unbalanced, and vice versa, purely because hybridization is less than 100% efficient. Further, if a second round of FISH is employed, in order to interrogate more chromosomes, the efficiency of the procedure decays, and it becomes inevitable that some misdiagnoses will be made (Wells et al., 2008). Munné et al. (2000b) estimated an average error rate of 6% based on studies of all blastomeres in a series of donated embryos, and 10% in real PGD situations; signal splitting, signal overlap, and incompletely penetrating probe may be the usual reasons. With particular reference to aneuploidy screening, Treff et al. (2010a) emphasize the potential for error due to poor spreading and fixation of cells. The most likely adverse outcomes would be wastage of normal embryos (diagnosed as monosomies) and diagnosis of embryos with a trisomy as normal. It is not surprising that centers that offer PGD for translocations usually recommend that it be followed up by conventional prenatal diagnosis to check that these sorts of error have not happened (Munné et al., 2002a). As discussed earlier, the technical capacity of FISH has been called into question, and SNPmicroarray may prove to be the superior methodology. MI CROARRAY AN ALYSI S
The first infant born following PGD-AS by arrayCGH was reported in 2001 (Wilton et al.), and the first array-CGH polar body study the following year (Wells et al., 2002). The case is now being made for the microarray approach, both for aneuploidy screening and for targeted imbalances due to a parental translocation (Treff et al., 2010a, 2010b, 2011a; Alfarawati et al., 2011; Fiorentino et al., 2011). The whole karyotype is in principle examinable. Microarray does have the advantage of not requiring specific or tailor-made probes: the methodology can be by hybridization of amplified DNA to a CGH- or SNP-based array. There is the disadvantage that the time taken for analysis often means that embryos have to be cryopreserved after biopsy, pending the result, although it may be that technical advances in vitrification of embryos, and in the laboratory methodology of single-cell microarray
analysis, will circumvent this question (Wells et al., 2008; Treff et al., 2009b; Vanneste et al., 2009a). An alternative molecular approach, in the specific case of a parent being a translocation carrier, is the measurement of short tandem repeat markers on the chromosomes in question, which can allow a quantitative assessment of the relevant segments, and thus distinction of euploidy versus aneuploidy (Traversa et al., 2010). In the case of polar body analysis, Landwehr et al. (2008) have developed a “rapid CGH” methodology, allowing diagnosis within a timeframe of 16 hours. Johnson et al. (2010b) have dealt with the problem of interpretation due to copy number variation by including parental DNA samples in the analytical procedure. As with FISH, the normal and the balanced carrier states cannot be distinguished.
Considerations of Embryology T HE PR O B L EM O F M OSA I CI SM A N D “CH AOT I C” EM B R YOS
The 6–10 cell embryo (the stage at which blastomere sampling is typically done) probably contains only one or two cells whose descendants will go on and form the inner cell mass, and thus, eventually, the embryo proper and the fetus. Chromosome studies on IVF embryos can reveal different chromosome constitutions in different cells, up to the point of “chaotic” embryos in which several cells each have a different aneuploidy, or collection of aneuploidies (Vanneste et al., 2009a and b), and as discussed earlier. It is not necessarily easy to guess (but intelligent guesses can be made), from the observed pattern of the different aneuploidies, what might have been the sequence of events at each individual mitosis that was able to lead to this eventual picture. Munné et al. (2002b) list these four main categories: diploid/polyploid mosaicism, chaotic mosaicism, mosaicism due to mitotic nondisjunction, and “split” mosaics with two cell lines that complement each other. A particular vulnerability may apply to these very early mitoses, before the necessary genes for cell-cycle checkpoint control have fully swung into action, and maternal cytoplasmic factors are still being relied upon (Hardy et al., 2002; Voullaire et al., 2002). Alternatively, or perhaps additionally, there may be a male factor involved, with impairment of the embryo’s centrosome function (Rodrigo et al., 2010). This could apply more particularly to Preimplantation Genetic Diagnosis •
435
cases of a severe spermatogenic defect, with a poorquality sperm bringing a poor-quality centriole to the embryo, given that the first few mitoses make use of the centriole that came with the sperm (Silber et al., 2003). But has the problem been overstated? As we discussed earlier, the power of the SNP-array may be bringing a clearer light, and exposing an inherent technical inadequacy in FISH methodology. Mosaicism can certainly happen, but it may be less frequent, and less chaotic, than we had thought in the early 2000s. THE N EED TO CO N S I DE R A F U LL RA NGE OF AB N O RMAL SEG R EGA N T OU TCOM ES
Postzygotic selection against aneuploid embryos has had little chance to have operated by day 3. Thus, in the case of translocation carrier parents, all possible segregant outcomes of a rearrangement may be encountered (see Fig. 5–4 and Table 5–2 in Chapter 5), and FISH probes will need to be selected accordingly. In the series of Ko et al. (2010), comprising some 1294 analyzed embryos, the fractions were as follows: alternate, 22%; adjacent-1, 23%; adjacent-2, 7%; 3:1, 25%; and 4:0, 2.5%; in the remaining 16% the segregation mode could not be determined. Somewhat similar fractions were reported in Mackie Ogilvie and Scriven (2002) and in Pujol et al. (2006); differences between the studies may likely reflect the particular nature of the translocations involved. A significant fraction with a balanced translocation complement, but a concomitant aneuploidy for one of the “common trisomic” chromosomes, namely 13, 16, 18, 21, and 22—8% of embryos in Pujol et al.—may warrant including an aneuploidy screen in the PGD analysis, and Treff et al. (2011a) have shown that this approach works. Some complete or partial autosomal monosomies, practically unknown otherwise, may be associated with “occult abortion” in the first 2 weeks post conception. Conn et al. (1999) describe PGD in the setting of a parental t(6;21), using a particular FISH strategy which covered the possibilities for chromosome 21 imbalances, and one embryo had a normal signal. This embryo was duly transferred, but with no more than a “biochemical pregnancy” resulting. In fact, this may have been interchange monosomy 6. And in the complex chromosomal rearrangement, the range of possible abnormality can be measured in hundreds; and yet, occasionally, a successful outcome is seen (Escudero et al., 2008). 436
•
P R E N ATA L D I A G N O S I S
GENETIC COUNSELING PGD is sufficiently complicated, not to mention expensive, that it will not usually be the first option for fertile couples wishing to avoid the birth of a child with a chromosomal disorder. High-risk scenarios might, however, warrant consideration sooner rather than later. Women may see access to the procedure as empowering, but equally, may find the process stressful; discarding an embryo with an unbalanced translocation, having had a child with that condition, may raise uncomfortable ambiguities (Karatas et al., 2010). For infertile couples (whether or not there is a chromosomal basis of the infertility) who require an IVF procedure to conceive, advice about a place for PGD-AS will need to be tempered by a continuing understanding of the differing relevance of meiotic and mitotic errors, and the factor of “chaotic mosaicism.” Equally, counselors will need to keep abreast of the continuing debate about the potential of FISH in diagnosing, or in overdiagnosing, mosaicism, and an emerging view that SNP-array may be the superior procedure. For those particular couples presenting for PGD, on the basis that one of them carries a chromosomal rearrangement, a number of points need to be raised.
The Reasons for Choosing Preimplantation Genetic Diagnosis as an Option Some couples may have had conventional prenatal diagnosis with successive terminations of pregnancies due to a high-risk translocation and be unwilling to face this prospect again. It may be difficult to distinguish a run of bad luck, with an optimistic outlook for the next pregnancy a realistic possibility, and therefore allowing the counselor to suggest a further natural attempt. Or the series of abnormal pregnancies may reflect a strong predisposition of that translocation to generate unbalanced gametes. Avoiding the possibility of termination following conventional prenatal diagnosis is, for those who have had that experience, a strong motivation (Lavery et al., 2002). T HE L I M I T ED S UCCESS R AT E
As discussed earlier, many IVF/PGD procedures do not produce the desired end result of a “take-home baby,” and the figures for PGD pregnancies are fairly similar to those applying to all IVF patients.
Thus, couples who would otherwise have no difficulty conceiving should weigh up the pros and cons of PGD and conventional prenatal diagnosis (Kanavakis and Traeger-Synodinos, 2002). Counselors seeing these couples need to be knowledgeable about all aspects of IVF and the PGD process, including an understanding of their local success rates. THE SPECIFIC GENETIC RISK
The figures provided elsewhere in this book largely relate to the risk for an unbalanced chromosome complement in either a liveborn child or at conventional prenatal diagnosis. Naturally, the risk that an embryo at PGD will be abnormal is substantially higher. From the ESHRE data for 2005–2006, the fraction of embryos from rcp carriers that were transferable was just 19%; thus, an 81% abnormality rate. For the robertsonian carrier, the figures differed somewhat between the sexes, with 28% of diagnosed embryos transferable from the female, and 39% from the male (Goosens et al., 2008). Bint et al. (2011) reviewed findings from gamete (polar body or sperm) analysis and in the preimplantation embryo in the two major categories of rob (13/14 and 14/21), with respect to all forms of imbalance, adjacent and 3:0. High fractions were seen in the female,
with similar figures for polar body and PGD: 33% unbalanced for the rob(13;14), and 40% for the rob(14;21). Lesser fractions of sperm and embryos were unbalanced from the male: 14% for the rob(13;14), and 11% of sperm and 5% of embryos with the rob(14;21). For recurrent rearrangements, risk data at the PGD stage may be pooled to give PGDspecific risk figures. Data relating to the common t(11;22)(q23;q11) are set out in Table 26–1 (and see also Table 5–2 in Chapter 5), and the rob figures just noted from Bint et al. apply to this chromosomal category. F OL LOW- UP I N T H E PR EG N A N CY
Understandably, some couples will be unenthusiastic about an invasive procedure that could possibly put at risk the pregnancy in which there has been so much investment (Meschede et al., 1998b). Nevertheless, couples need to be aware that chromosomal PGD cannot provide a “guarantee,” albeit that the misdiagnosis rate, for whatever reason, is very low, when good-quality embryos are transferred (Wilton et al., 2009). Prenatal diagnosis should be offered. Ultrasonography may be an acceptable, if imperfect compromise, only proceeding to CVS or amniocentesis if anomalies are detected. Maternal serum
Table 26–1. Risks of Generating Balanced and Unbalanced Embryos for Seven Carriers of the Common t(11;22)(q23;q11) NO. OF EMBRYOS SEGREGATION MODE
ALTERNATE
ADJ-1
ADJ-2
3:1
4:0
OTHER*
Van Assche et al. case 2 (male) Munné et al. case E (male) Mackie Ogilvie and Scriven (male) Average proportions (male)
9 1 9 33%
3 2 4 16%
2 0 1 5%
1 0 0 2%
0 0 1 2%
19 5 0 42%
Van Assche et al. case 1 (female) Iwarsson et al. case 5 (female) Ibid. case 11 (female) Mackie Ogilvie and Scriven (female) Average proportions (female)
0 5 2 0 23%
1 3 0 0 13%
0 0 0 0 0
2 3 2 2 30%
0 0 0 1 3%
0 4 5 0 30%
Notes: Embryos were studied at PGD and subsequently, for untransferred embryos, at rebiopsy with as many cells as could be analyzed. These data are very scant but show the beginnings of how such information may eventually come to be accumulated. It is interesting that the favoring of alternate segregation in the male seen here is not reflected in the sperm data of the single heterozygote listed in Table 5–1 (see Chapter 5), in which adjacent-1 is the predominant mode. See also Table 5–2. *Unbalanced but mode not analyzable; mosaicism; chaotic mosaicism; polyploidy. Adj-1, adjacent 1; adj-2, adjacent-2. Sources: Van Assche et al. (1999), Munné et al. (2000b), Iwarsson et al. (2000), Mackie Ogilvie and Scriven (2002).
Preimplantation Genetic Diagnosis •
437
screening (p. 410) offers a further possibility, although it is necessary to take account of the fact that PAPP-A levels are less in IVF pregnancies, which might otherwise have been interpreted as an increased risk for trisomy 21 (Amor et al., 2009).
NATUR E MAY IN TER V E NE
A natural pregnancy may be achieved while the couple waits for the IVF/PGD preparations to be made. For example, the adjacent-2 karyotype shown in Figure 5–10 (see Chapter 5) came from culture of the products of conception of this couple’s third miscarriage, and no normal pregnancies, the woman being a t(13;16) carrier. The outlook did not seem very promising, and plans were being put in place for IVF; but they then reported a naturally conceived pregnancy, in which amniocentesis showed a 46,XY karyotype.
438
•
P R E N ATA L D I A G N O S I S
T HE CH I L D R EN R ES ULT I N G
Children born from a PGD pregnancy, a minority group among the large numbers worldwide of “IVFlings,” appear to be of essentially normal health (Banerjee et al., 2008; Desmyttere et al., 2009). They may be born slightly earlier, and be a little lighter, than normally conceived babies. These risks, and a risk for perinatal death, may be elevated in the case of a multiple pregnancy (Liebaers et al., 2010). Perhaps unsurprisingly, for a child in whom so much has been invested, PGD infants score well on a scale of “warmth–affection” (measured by observing how infants may be cuddled and kissed, and how positively and kindly spoken to). We can anticipate longer-term studies on the health and development of these “PGD children,” in the fullness of time (a present lack whereof is “deplored” by Liebaers et al.). The risk for Beckwith-Wiedemann syndrome in IVF babies generally is noted on p. 369.
27 chromosome abnormalities detected at prenatal diagnosis
THE MAIN FOCUS of chromosomal prenatal diagnosis has been upon trisomy 21, usually in the context of older childbearing age or of an increasedrisk screening test. Trisomy 21 does remain, for most women and couples, the prime concern—the condition that most people are aware of—but with the modern ability to detect subtle imbalances on direct testing at chorionic villus sampling and at amniocentesis, the great majority of chromosomal imbalances are, in principle, diagnosable. And the increasing sophistication of screening methodologies (Chapter 24) has meant that a substantial fraction of chromosomally abnormal pregnancies are, these days, diagnosed prenatally.
DECISION MAKING FOLLOWING PRENATAL DIAGNOSIS OF A CHROMOSOMAL ABNORMALITY To some extent, the possibility of “other abnormalities besides Down syndrome” should have been raised at counseling before prenatal diagnosis, in
women presenting for amniocentesis or CVS as a routine discretionary procedure, or following an increased-risk assessment from screening, or indeed in whom a fetal abnormality is already known, on the basis of ultrasonography. But when a chromosomal abnormality is actually discovered, it is of course necessary to discuss in detail with the couple the implications of this particular abnormality and to help them decide on a suitable course of action. Outlines of the clinical consequences of these abnormalities follow, to serve as a basis for the decisions that these women and couples need to make. In transmitting the information, the counselor is obliged to be clear and accurate about the particular abnormality, and to take care that the couples’ autonomy in the decision-making process is not compromised. A decision for or against termination is the immediate one to be made. Some years ago, but their view remains valid, Engel et al. (1981) listed these factors influencing the parents’ decision: their philosophy of life, their religious views, their socioeconomic status, and whether this was a •
439
first or wanted pregnancy or a later, unplanned pregnancy. Unsurprisingly, the severity of the condition influences decision making. Drugan et al. (1990) found that 93% of parents having a prenatal diagnosis with a “poor prognosis” (autosomal trisomy, unbalanced translocation, 45,X with major anomalies on ultrasonographic examination) chose pregnancy termination, while only 27% of parents given a “questionable prognosis” (sex chromosome aneuploidy, 45,X with normal ultrasonography, de novo apparently balanced translocation or inversion) took this course. Shaffer et al. (2006a) undertook a large retrospective review (1983–2003), analyzing parental decisions in 816 prenatal diagnoses of a major aneuploidy, at a San Francisco clinic. Termination was chosen in 86% of autosomal trisomy, and in 60% with a sex chromosome aneuploidy. Of the latter, the rates of termination increased progressively from XXX (40%), XYY (57%), 45,X (65%), to XXY (70%), in parallel with a perceived severity of phenotype. The rates did not differ significantly over the 21-year period, to the slight surprise of these authors. Drugan et al. make the interesting observation that ultrasound visualization of fetal defects “in a society dominated by the television screen” can be useful in helping parents better grasp the implications of the diagnosis; although seeing an image of the actual fetus can also sharpen the ethical dilemma inherent in confronting the possibility of termination of a pregnancy (Williams et al., 2005). TRI SO MY 2 1
Skotko et al. (2009) emphasize the need for the person conveying the news of a Down syndrome (DS) result to be well informed, whether that be a counselor, obstetrician, or other health professional (and this qualification scarcely confined to a diagnosis of DS). Ideally, the news should be given in person; but where that is not feasible, a phone call should be at a prearranged time. Parents who decide to continue a trisomy 21 pregnancy, versus those who have chosen termination, would presumably come from different points of view. Skotko et al. note that contact with a DS support group might be useful for some couples in deciding the fate of a DS pregnancy, although they do observe that few studies have assessed the views of those who have terminated a trisomic pregnancy, from whom the other face of the decision could be given a hearing. A study of health professionals in Finland showed some 440
•
P R E N ATA L D I A G N O S I S
inconsistency in comparing the points of view of midwives and public health nurses with the options available to their patients (and the acknowledgment made that this difference could be seen as a healthy sign) ( Jallinoja et al., 1999). Thus, most (79%) of these midwives and nurses agreed that all pregnant women should be offered a screening test, although only 44% personally accepted the concept of genetic abortion. An acceptance of abortion correlated with education and with a professional experience with DS patients. In the United States, Britt et al. (2000) studied 142 women who had had a prenatal diagnosis of trisomy 21, seen in Detroit over the period 1989–1998. Those who had already had children, and where the diagnosis of trisomy 21 was made earlier in the pregnancy, were more likely to choose termination. SE X CH R O M OS O M E A B N O R M A L I TY
The grayest area is sex chromosome aneuploidy, and views have been changing somewhat over recent decades, at least in the West, generally in the direction of a more conservative response to the news of a chromosomal abnormality (Christian et al., 2000; Linden et al., 2002; Boyd et al., 2011). In Denmark in 1986, Nielsen et al. reported that approximately 80% of prenatal diagnoses of sex chromosome aneuploidy at that time were followed by the choice of abortion. In an English/Finnish study from the same period, termination (in about 60% overall) was more likely to be chosen in the case of the XXY and 45,X karyotypes, by younger parents with fewer previous children, and in all cases in which an ultrasonographic defect was identified (Holmes-Siedle et al., 1987). From a large survey of centers in five European countries, covering the years 1986 to 1997, the rate of choice of termination with respect to XXY was 44% (Marteau et al., 2002). In a German study over a similar period, termination was chosen by a much smaller fraction, only 13%, among parents who had been given a prenatal diagnosis of 47,XXX, 47,XXY, or 47,XYY (in contrast, just 2% of parents at the same clinic decided to continue a pregnancy with trisomy 21) (Meschede et al., 1998c). This may in part have reflected the practice of this clinic to emphasize the point that “the mean global IQ of around 90 falls well within the normal range and is compatible with a productive and socially welladjusted life.” In more recent years, a similar reduction in the choice of termination has been seen in France (Brun et al., 2004). A quite different
experience comes from China, however (Liao and Li, 2008). Almost all pregnancies with a fetal diagnosis of sex chromosome abnormality are terminated. In considerable part, this may reflect the influence of the “one-child policy,” with couples wanting the best outlook for their one and only child. In the specific case of 45,X and variants, from 19 registries in 10 countries across Europe from 1996 to 1998, 79% of parents chose termination if morphological abnormality, and in particular cystic hygroma, had been seen on ultrasonography, versus 42% in which the diagnosis had not been led into by an ultrasonographic defect (Baena et al., 2004). Parental attributes may be (as in many respects!) important in influencing the eventual outcome of these children. In the experience of the Denver group, for example, the parents choosing prenatal diagnosis were often of higher socioeconomic status, and the children of those who had made conscious decisions to continue the pregnancy, following the discovery of a sex chromosome abnormality, had generally done better than those identified in population newborn surveys (Linden and Bender, 2002). The way in which information is given has an important impact, and counselors need to be well aware of the weight that parents, in some emotional turmoil at the news they have just received, may put upon the news given them. Consider the example of 47,XXY Klinefelter syndrome. In the European survey mentioned earlier, Marteau et al. (2002) assessed responses to the prenatal diagnosis of XXY when counseling had been given by obstetricians, pediatricians, midwives, health visitors, or genetics specialists. Women counseled solely by genetics specialists were more than twice as likely (relative risk = 2.4) to continue the pregnancy versus those counseled either by other professionals or by other professionals along with a geneticist. It seems probable that these differences may reflect the style of counseling. Marteau et al. (1994) make the following distinctions in counseling types: nondirective counseling (“try to be as neutral as possible, covering both positive and negative aspects”), directive counseling for termination (“encourage termination” or “try to be as neutral as possible but overall convey more negative than positive aspects of the condition”), or directive counseling against termination (“encourage parents to carry to term” or “try to be as neutral as possible but overall convey more positive than negative aspects of the condition”). The desirability for a consistent approach, with access to accurate information, is to be emphasized,
as is—of course—the requirement to enable women’s choices to be well informed in the broadest sense, and for the counseling to be nondirective (Abramsky et al., 2001; Marteau and Dormandy, 2001; Linden et al., 2002). Beyond the clinic, there are support groups, public information resources, and talking with other parents, as means to become further informed about the implications of a sex chromosome abnormality (in the short period of time during which a decision must be made), and Linden et al. (2002) note the pros and cons of taking these paths; as noted earlier with respect to trisomy 21, the views of those who had previously chosen to terminate a pregnancy are less readily accessible. The prime responsibility for putting couples in the best position to make an appropriate decision lies with the counselor. As for subsequently informing the children from the pregnancies that are continued, Sutton et al. (2006) emphasize the importance of telling them of their sex chromosomal diagnosis (specifically, Turner syndrome), and its implications, in a timely and sensitive manner, and of not “keeping secrets.” SU B M I CR OS CO PI C “ M I CR OA R R AYLE V EL ” R EA R R A N G EM EN T
Microarray analysis applied to prenatal samples is capable of detecting imbalance practically at the level of the operator’s choice, according to which particular commercial or in-house chip is used (Rickman et al., 2006; Shaffer and Bui, 2007; Shaffer et al., 2008; Coppinger et al., 2009; Van den Veyver et al., 2009) (and see Chapter 2). Furthermore, the analysis can be performed on small amounts of material, and results may be obtained with a 48-hour turnaround. As noted on p. 418, microarrays can increase the prenatal diagnostic pickup, following discovery of an ultrasound defect, by 2%–4%. The other side of this two-edged sword is the fact that some microimbalances are not pathogenic and may simply reflect “copy number variation.” Indeed, one commentator has written, somewhat provocatively, that prenatal array testing is likely “to produce a flood of information that is overwhelming, anxietyproducing, inconclusive and misleading” (Shuster, 2007). A response to this is to target the array: ask the right question, if we want a useful answer. That is, we can interrogate only those chromosomal segments for which precedent exists as being causative of an abnormal phenotype, assessing in particular the known microduplication/microdeletion syndromes, Chromosome Abnormalities
•
441
along with subtelomeric and pericentromeric regions (South and Lamb, 2009). Or, a little more broadly, to target, in addition, gene-dense regions, on the assumption that these might more plausibly be, when duplicated or deleted, pathogenic. Using these targeted approaches, the results of unclear clinical significance are minimal and are not substantially different than those found by routine cytogenetics (Shaffer et al., 2008; Coppinger et al., 2009a; Van den Veyver et al., 2009). Counselors who see these patients need to be quite au fait with the interpretations and to maintain close liaison with expert scientists in the field (and see also p. 274).
MOSAICISM: CONFINED, CONSTITUTIONAL, AND PSEUDO Mosaicism is the bane of cytogenetic prenatal diagnosis. Most times, it turns out to have been a false alarm, and the mosaicism in villus tissue or amniocytes does not reflect a true constitutional mosaicism of the embryo. This is a problem for the laboratory to resolve, inasmuch as they are able. We may list these categories: confined placental mosaicism, true constitutional mosaicism, and pseudomosaicism. A chromosomally abnormal cell line may exist only in extra-embryonic tissues (chorion, amnion), and the embryo is 46,N. This is confined placental mosaicism (CPM). CPM is encountered at CVS rather than at amniocentesis. It is uncommon that an observation of apparent CPM at CVS reflects a true constitutional mosaicism of the fetus. Stetten et al. (2004) reviewed a series of 4000 CVS studies done over the period 1998–2003, in which 29 cases (0.7% of the total) of CPM were defined. Testing of the newborns revealed two as having (low-level) true mosaicism. A long-term follow-up study (Amor et al., 2006) is noted later. In “pseudomosaicism,” the embryonic and extra-embryonic tissues are all 46,N, and the abnormality arose during tissue culture in vitro (“cultural artifact”). Considerable discussion follows: but at the outset, we should emphasize that true mosaicism of the fetus is infrequently observed, and that the majority of mosaicism identified at prenatal diagnosis, more especially at CVS, does not presage an abnormal baby. It is important to keep this perspective in talking with parents (according to the particular attributes of the mosaicism, as we go on to discuss) and to avoid causing any more anxiety than that which, inevitably, an “abnormal” result brings. 442
•
P R E N ATA L D I A G N O S I S
Applied Embryology Interpreting mosaicism obliges an understanding of the earliest events of development of the conceptus (Bianchi et al., 1993; Robinson et al., 2002). The zygote undergoes successive mitoses to produce a ball of cells (morula) (Fig. 27–1,1). The morula then cavitates to produce an inner cyst, and it becomes the blastocyst (this is happening at the beginning of the second week post conception) (Figs. 27–1,2). The outermost layer of the blastocyst is comprised of trophoblast, and this tissue becomes the outer investment of the chorionic villi. The inner cell mass protrudes into the blastocystic cavity, and this will give origin to the embryo. It comprises two different cellular layers, the epiblast and the hypoblast. In a 64-cell blastocyst, most cells are trophoblasts, the inner cell mass comprises about 16 cells, within which only about 4 (epiblast) cells will give rise to the embryo itself. The hypoblast forms the spherical primary yolk sac (whose roof is, transiently, the ventral surface of the embryo). The primary yolk sac gives rise to the extra-embryonic mesoderm, sandwiched between itself and the outer cytotrophoblast, thus producing a three-layered sphere. The mesodermal cells now invade the blastocystic cavity (Fig. 27–1,3), and this mesodermal mass is in turn cavitated to produce the extra-embryonic celom, such that there are outer and inner layers of extra-embryonic mesoderm. The outer layer, underlying the trophoblast, gives rise to the mesenchymal core of the chorionic villus, and the inner layer becomes the outer (mesodermal) surface of the amniotic membrane. The amniotic cavity enlarges at the expense of the extra-embryonic celom (Figs. 27–1,5, 27–2) and eventually obliterates it (by the end of the first trimester), with the mesodermal layer of the amnion fusing with the mesodermal layer of the chorion. The epiblast gives rise to the amniotic cavity, the floor of which is the “dorsal” (ectodermal) surface of the embryo, and its roof is the amnion, these being continuous at their margins. Thus, the embryonic integument and the inner surface of the amniotic membrane—which are the source of the embryonic and amniotic epithelial cells present in amniotic fluid—have the same lineage. At the beginning of the third week, the primitive streak arises from the epiblast, and this in turn gives origin to both endoderm and intraembryonic mesoderm. Endoderm gives origin, among other tissues, to urinary tract and lung epithelia, desquamated cells from which
Yolk sac
Hypoblast
Inner cell mass (1) Morula
Epiblast (2) Blastocyst Trilaminar embryonic disk
Extraembryonic celom
Amniotic cavity (3) Blastocyst
Trophoblast
(4) Pre-Embryo
Ectoderm Mesoderm Mesoderm (extra-embryonic) (embryonic)
Endoderm
Amniotic cavity
Chorion
Gut Amniocentesis needle
Amnion
Extraembryonic celom
Body stalk
CVS needle
Yolk sac
Trophoblast Mesodermal core
Chorionic Villus
(5) Embryo
FIGURE 27–1 Developmental origins of tissues sampled at prenatal diagnosis (simplified). (1) Morula, 3–4 days post conception, a sphere with trophoblast cells at its surface. (2) Cross-section of blastocyst at beginning of second week, showing outer rim of trophoblast, the inner cell mass comprising epiblast (orange) and hypoblast (yellow), and the yolk sac cavity lined by an inner rim of cells of hypoblastic origin. (3) Blastocyst toward end of second week. The hypoblastic cells of the yolk sac have given rise to extra-embryonic mesoderm. Lacunae are beginning to appear in this mesoderm, and these will coalesce to form the extra-embryonic celom. (4) “Pre-embryo.” The amniotic cavity and the yolk sac bound the dorsal and ventral surfaces of the embryonic disk. The extra-embryonic celom has now cavitated the extra-embryonic mesoderm. Note that the embryonic mesoderm (middle layer of the trilaminar embryonic disk) arises from the epiblast, and thus it has a different lineage from the extra-embryonic mesoderm. (5) Composite embryo/early fetus. (Rotation has reversed the relative positions of the yolk sac and amniotic cavity.) The three embryonic tissue types (ectoderm, mesoderm, endoderm) all had origin from the epiblast, as did the amniotic epithelium. Epithelial cells from the embryo’s ectodermal surface are shed into the amniotic cavity, as also are amniotic epithelial cells (both these tissues shown orange). Cells from endodermal derivatives (respiratory and urinary tracts, which originate from the gut, shown in yellow) pass into the amniotic cavity. Chorionic villi comprise mesenchymal core (of extra-embryonic mesodermal origin), gloved by trophoblast. Extra-embryonic and embryonic mesoderms are continuous at the body stalk, albeit that some embryonic mesodermal cells may then migrate into the amniotic mesoderm (Robinson et al., 2002). See also separate color insert.
FIGURE 27–2 Ultrasound picture of embryo at 10–11 weeks gestational age, very close to actual size (note cm. markers at right). Note amnion (A), amniotic cavity (AC), extra-embryonic celom (EC), umbilical cord (U), “physiological omphalocele” (O), yolk sac (Y), and placenta (P). (Courtesy H. P. Robinson.) The relative positions of embryo and other structures are similar to the depiction in the drawing in Figure 27–1, part 5.
contribute to the cellular population of amniotic fluid. Albeit that the extra- and intraembryonic mesoderms have different origins, there may be migration of some intraembryonic mesodermal cells into the (extra-embryonic) amniotic mesoderm. Cells from the latter add a minor fraction to the population of amniocytes, but have a proliferative advantage, and may come to comprise most of the cells present following in vitro culture. Amniocentesis is, therefore, a procedure that samples cells having origin from the epiblast of the inner cell mass, and these cells rather closely reflect the true constitution of the embryo. Chorionic villus sampling, on the other hand, samples more distantly related cells: trophoblast cells (direct and short-term culture), which were the first lineage to differentiate from totipotent cells of the morula, and villus core cells (long-term culture), which reflect the more recently separated lineage of the extra-embryonic mesoderm. The differing origins of tissues sampled by different means are set out in Figure 27–3.
Mechanisms of Mosaicism Mosaicism may involve aneuploidy for an intact chromosome or for an abnormal chromosome, along with a normal cell line. Two broad formats may apply: first, a mitotic error in an initially normal conceptus which gives rise to an abnormal cell line; 444
•
P R E N ATA L D I A G N O S I S
or, second, an initially abnormal conceptus, typically due to a meiotic error, with a subsequent mitotic event generating a normal cell line (Fig. 3–8 in Chapter 3). The distribution of the normal and the abnormal cell lines in the fetus and the placenta depends upon the time and the place of the abnormal mitotic event. If, for example, a trisomic conceptus is “rescued” by the generation of a normal cell line, at a very early stage, in a cell that is going to give rise to the inner cell mass and to some of the extrafetal tissues, then the embryo may be 46,N, and the placenta will show mosaic trisomy. If rescue occurred at a later stage, the placenta might be entirely trisomic, with a mosaic trisomy of the fetus. These and other possible combinations are depicted in Figure 27–4. The eventual phenotype will be influenced by the tissue distribution of the cell lineages that contain the trisomic chromosome, and the normal:trisomic proportions in various tissues. The potential for widely differing tissue distributions of the different cell lines may confound interpretation at prenatal diagnosis. Consider the case of Jewell et al. (1992). A dup(12) chromosome was present in 87% of amnion cells, 60% of fetal blood, but only 2% of chorionic villi and in 0% of chorionic membrane. Kingston et al. (1993) provided a similar remarkable (and disconcerting) example. Amniotic fluid cells had 3% with an SMC, a sample of fetal blood showed all cells 46,N, and several tissues
FIGURE 27–3 Diagram of cell lineages arising from differentiation in the very early conceptus. The fertilized egg (1) produces a trophoblast precursor (1b) and a totipotent stem cell (2), which in turn forms another trophoblast precursor (2b) and a stem cell (3) that produces the inner cell mass. The inner cell mass divides into stem cells for hypoblast (3b) and epiblast (4). The epiblast cell(s) (5) produce embryonic ectoderm and primitive streak, and the latter is the source of embryonic mesoderm and endoderm. The cell lineages sampled at various prenatal diagnostic procedures are indicated at right. E, epiblast; H, hypoblast; P, primitive streak; Y, yolk sac. (From D. W. Bianchi et al., 1993, Origin of extra-embryonic mesoderm in experimental animals: relevance to chorionic mosaicism in humans, American Journal of Medical Genetics 46:542–550. Courtesy D. W. Bianchi; reproduced with the permission of Wiley-Liss.) This construction is to be compared with that of Kennerknecht et al. (1993b), in which three postzygotic mitoses occur, producing eight totipotent cells, before the cells begin to take on their tissue identities. Robinson et al. (2002) propose a further variation, with some cells of the embryonic mesoderm migrating into the (otherwise extra-embryonic) mesodermal layer of the amnion.
taken post termination had various fractions of mosaicism, including brain with 88% of cells aneuploid. Stankiewicz et al. (2001c) report an infant with the nonmosaic karyotype 46,X,der(Y)t(Y;7) (p11.32;p15.3) causing a 7p trisomy syndrome,
following the CVS diagnosis of very low-grade mosaicism 46,XY[49]/46,X,der(Y)t(Y;7)[1], and yet with nonmosaic 46,X,der(Y)t(Y;7) at amniocentesis. These observations point to an early postzygotic origin of the translocation in an initially 46,XY Chromosome Abnormalities
•
445
FIGURE 27–4 Types of mosaicism of the fetal-placental unit. Fetus depicted enclosed in its sac at right, with the chorionic villi comprising the placenta to left. Gray areas indicate an aneuploid cell line; white areas indicate karyotypic normality. In reality, the distributions of the two cell lines is unlikely to be as clear-cut as is shown here. In the examples showing placental mosaicism, the path taken by the sampling needle will determine whether the abnormality is detected or missed at chorionic villus sampling. The cartoon of the fetus, sac, and placenta is close to the form and about two-thirds of the size that actually exists at 10 weeks 0 days (gestational age as measured clinically, dated from the last menstrual period), when crown-rump length is around 30 mm.
conceptus, apparently affecting the entire inner cell mass but only a very small minority of trophoblasts. These three cases, admittedly exceptional, are instructive in emphasizing that the proportions of abnormal cells in one tissue can not necessarily be taken as indicative of proportions elsewhere. 446
•
P R E N ATA L D I A G N O S I S
Laboratory Assessment of Mosaicism The resolution of mosaicism in the cytogenetics laboratory and in its clinical interpretation can differ for CVS and amniocentesis, and we will consider them separately. In terms of the laboratory result, we
can apply to both CVS and amniocentesis the concept of different levels of in vitro mosaicism, originally developed for amniocentesis by Worton and Stern (1984), and refined by Hsu et al. (1992) and Hsu and Benn (1999), as follows: Level I. A single abnormal cell is seen. With near certainty this is cultural artifact, and is thus pseudomosaicism, and this may be resolved after additional workup as set out in Table 27–1. The laboratory would not usually report the single cell observation, if the analysis of additional cells failed to confirm the abnormality.1
Level II. Two or more cells with the same chromosomal abnormality in a dispersed culture from a single flask, or in a single abnormal colony from an in situ culture (i. e., possibly or probably just a single clone). Some would also include the observation of two or more colonies from the same in situ culture. The abnormality is not observed in multiple colonies from other independent cultures. This form of mosaicism is almost always pseudomosaicism. It would not usually be reported to the physician if additional workup failed to confirm the trisomy, but it may be reported if additional studies were inadequate, if fetal anomalies had been identified, or in
Table 27–1. Guidelines for Workup for the Elucidation of Possible Amniocyte Pseudomosaicism/Mosaicism FLASK METHOD
A. Indications for Extensive Workup (1) Autosomal trisomy involving a chromosome 21, 18, 13; or 2, 5, 8, 9, 12, 14, 15, 16, 20, 22 (SC, MC) (2) Unbalanced structural rearrangement (MC) (3) Marker chromosome (MC) B. Indications for Moderate Workup (4) Extra sex chromosome (SC, MC) (5) Autosomal trisomy involving a chromosome 1, 3, 4, 6, 7, 10, 11, 17, 19 (SC, MC) (6) 45,X (MC) (7) Monosomy (other than 45,X) (MC) (8) Marker chromosome (SC) (9) Balanced structural rea (MC) C. Standard, No Additional Workup (10) 45,X (SC) (11) Unbalanced structural rea (SC) (12) Balanced structural rea (SC) (13) Break at centromere with loss of one arm (SC)
IN SITU METHOD
(1) Autosomal trisomy involving a chromosome 21, 18, 13; or 2, 5, 8, 9, 12, 14, 15, 16, 20, 22 (SCo, MCo) (2) Unbalanced structural rearrangement (MCo) (3) Marker chromosome (MCo) (4) Extra sex chromosome (SCo, MCo) (5) Autosomal trisomy involving a chromosome 1, 3, 4, 6, 7, 10, 11, 17, 19 (SCo, MCo) (6) 45,X (SCo, MCo) (7) Monosomy (other than 45,X) (SCo, MCo) (8) Marker chromosome(SCo) (9) Balanced structural rea (MCo) (10) Unbalanced structural rea (SCo) (11) Balanced structural rea (SCo) (12) Break at centromere with loss of one arm (SCo) (13) All single-cell abnormalities
Notes: Criteria for extensive (A.), moderate (B.), and standard (C.) workup: A. Forty cells (20 cells from each of two flasks, excluding those cells analyzed from the culture with the initial observation of abnormality), or 24 colonies (excluding those colonies analyzed from the vessel with the initial observation). B. Twenty cells (from the flask without the initial observation), or 12 colonies (from vessels without the initial observation). C. Twenty cells (10 from each of two independent cultures), or 15 colonies (from at least two independent vessels). MC, multiple cells (single flask); MCo, multiple colonies (single dish); Rea, rearrangement; SC, single cell (single flask); SCo, single colony (single dish). Source: From Hsu and Benn (1999).
1 An exception may be mosaicism for an isochromosome, as a handful of reports have demonstrated true mosaicism in the context of a single abnormal cell at prenatal diagnosis (see later).
Chromosome Abnormalities
•
447
Table 27–2. Probabilities of Survival to 1 Week, 1 Month, and 1 Year, for Liveborn Infants with Trisomies 13, 18, and 21
Trisomy 13 Trisomy 18 Trisomy 21
1 WEEK
1 MONTH
1 YEAR
0.42 (0.06–2.93) 0.52 (0.07–3.69)
0.20 (0.03–1.41) 0.30 (0.04–2.10) 0.98 (0.97–0.99)
0.03 (0.00–0.22) 0.03 (0.00–0.19) 0.95 (0.93–0.96)
Note: 95% confidence limits in brackets. Source: Vendola et al. (2010).
the case of certain chromosome abnormalities which are well recognized as existing in the mosaic state (e.g., trisomy 16; see later). A course of action to resolve the issue cytogenetically, in the case of amniocentesis, is given in Table 27–1. Level III. Two or more cells with the same chromosome abnormality, distributed over two or more independent cultures. Level III is likely to reflect a true mosaicism, and the cytogeneticist will report this finding immediately. (Some allow level III to include more than one colony in only a single flask, although this could be an “over-interpreted level II” if two colonies in the one flask had arisen from a single cell whose progeny migrated and established separated clones.) The distinction may not be quite as clear as this in practice, but this is a useful working definition. The mathematics of sampling comes into the picture: how many cells need to be looked at, in order to establish what level of confidence that the possibility of mosaicism of what extent can safely be disregarded? Tables have been derived to assist in answering this question (Hook, 1977; SikkemaRaddatz et al., 1997a). Inevitably, low-level mosaicism will, on rare occasions, be missed. Given the reality that only a limited number of cells can be karyotyped, the statistics will sometimes conspire against the cytogeneticist, and only normal cells will be examined. This has to be accepted: the test is not perfect. For example, de Pater et al. (2003a) describe their experience in reporting a normal result from amniocentesis, but in due course the child proving to be a r(12) mosaic, with a high level of 50% on blood. Critically reviewing their procedures, and indeed being able to see the ring chromosome when archived material from the amniocentesis was restudied, they nevertheless drew the conclusion that their original analysis has been appropriately performed. A similar example, with respect to a CVS 448
•
P R E N ATA L D I A G N O S I S
case, is noted later, in the section on 47,+i(5p). In CVS, the exposure to error may relate to the part of the placenta the sampling needle happens to traverse. NE WER M O L ECUL A R M ET H O D O LO G I ES A ND M OSA I CI SM
Mosaicism may be detected with reasonable efficiency in qualitative fluorescent polymerase chain reaction (QF-PCR). In one large retrospective study, Donaghue et al. (2005) reviewed 8983 amniocentesis and CVS samples, from which 18 cases with mosaicism were identified. More (12) were detected by QF-PCR than by karyotyping (8), although neither approach picked up all. By their reckoning, a tissue load of 15% or more abnormal cells would allow detection of mosaicism by QF-PCR. Concerning microarray, Ballif et al. (2006) tested the system, using experimental dilutions of a 46,XY sample with a 47,XY,+21 sample, in order to mimic trisomy 21 mosaicism; and they demonstrated that mosaicism of 20% or greater could confidently be identified. Similarly, Cross et al. (2007) set up mock samples from normal and trisomy 8 fibroblasts, and, by analyzing the extracted DNA with a 50K SNP array, they established that, down to a 20% level, mosaicism was readily recognized, but fading out at about 10%. In terms of actual experience, Filges et al. (2011) detected three cases of differing forms of mosaicism among 80 high-risk pregnancies tested by array-CGH, and show, unsurprisingly, that the same dilemmas can arise with confined placental mosaicism as seen in conventional karyotyping. PRED I CT I O N O F PH EN OTYPE I N A N I N D I V I D UA L CA SE
Elegant theorizing notwithstanding, the pragmatic observations from published cases in the literature provide the mainstay of the advice that the counselor
may offer the parents in an individual case. (Large series are better than single case reports, which are better than anecdote.) Are mosaicisms for some particular chromosomes, or types of aberration, of more concern than others? What is a low enough level of mosaicism, if any such exists, to have a degree of confidence that the child will be physically and mentally normal? We set out later summaries of the recorded examples from the literature, none of which necessarily provide a firm answer, but which may serve as the basis for discussion and counseling. The numbers in some are very small. Another difficulty with these observational data is that, for the most part, the window of assessment of the child’s phenotype was confined to the neonatal period. Of course, many children who are eventually diagnosed with significant handicap may have been well grown and morphologically normal at birth, with normal functional neurology (inasmuch as this may be assessed in a baby). On the other hand, it is possible to overdiagnose problems in babyhood, as a child who subsequently develops normally may prove (Warburton, 1991; Joyce et al., 2001). An important concern in mosaicism is that a cell line inaccessible to analysis—specifically, in the brain2—might contain the abnormal chromosome, notwithstanding a normal karyotype in the postnatal tissues which are normally examined, namely, blood and possibly skin. If so, cognitive functioning could be compromised. Those few reports that include follow-up data for some years into childhood (Baty et al., 2001; Amor et al., 2006) are therefore most valuable. Nevertheless, no certainty can be offered, recognizing that every case of mosaicism will be unique, in terms of the extent and qualitative tissue distribution of the abnormal lineage.
CHORIONIC VILLUS CULTURE AND MOSAICISM, INCLUDING CONFINED PLACENTAL MOSAICISIM CVS mosaicism is detected in less than 1% of procedures at the 10- to 11-week mark. (A previous figure
of ~2% applied when direct/short-term culture was widely used, which favored assessment of the cytotrophoblast; but few laboratories nowadays do this analysis.) Mosaicism from an early mitotic error in a single cell can give rise to confined mosaicism (confined to placenta, or to fetus) or to generalized mosaicism (present in both fetus and placenta), according to the destined lineage of that cell; the broad range of possibilities is shown in Figure 27–4. Depending upon the timing and site of the event producing the mosaic state, the karyotypes observed at CVS will vary. The extreme form is complete discordance, with a nonmosaic 46,N karyotype in fetus and nonmosaic aneuploidy in CVS, or vice versa.3 Clearly, an important distinction to make, inasmuch as it is possible to do so, is between a mosaicism confined to the placenta (CPM), and causing little or no compromise of its function, versus the presence of an aneuploid cell line extending into the fetus, plus or minus an important effect upon the ability of the placenta to support fetal development. Follow-up amniocentesis is certainly advisable: a normal result, which is very often what eventuates, will substantially provide reassurance that the aneuploidy did not involve fetal tissue. However, as a broad-brush estimate, Daniel et al. (2004) assess that on the order of 10% of CVS mosaicism for certain “rare chromosomes”4 interpreted as CPM, may in fact reflect a cryptic fetal mosaicism, that would not be detected at follow-up amniocentesis, and which might or might not have important phenotypic consequence (see later section on “Prognosis”). Our focus here is on confined placental mosaicism. One classification of CPM is as follows: type I, aneuploidy confined to cytotrophoblast (recognized only at direct/short-term analysis); type II, aneuploidy confined to villous stroma; and type III, an aneuploid cell line in both cytotrophoblast and stroma. ORI G I N O F T R I S O MY I N CO N F I N ED PLACENTAL MOSAICISM
Robinson et al. (1997) studied 101 cases in which CPM had been identified at CVS, seeking to establish
2 While we do not overlook the astonishing suggestion of Westra et al. (2008) that mosaicism in individual brain neurons may be a normal phenomenon, it is scarcely debatable that genomic imbalance due to a specific chromosomal defect, in a fraction of cells of the cerebral cortex, would in some wise affect intellectual functioning. 3 CPM is the main, but not the only cause of discrepancy between the CVS and fetal/child karyotypes. One very rare explanation is that there was a resorbed co-twin with a different karyotype, with the sampling instrument having traversed its placental remnant (Tharapel et al. 1989). 4 In this study, chromosomes 5, 8, 9, 10, 11, 12, 14, 15, and 16.
Chromosome Abnormalities
•
449
correlates of the origin of the trisomy. Some CPM trisomies are usually of mitotic (somatic) origin, the zygote having been 46,N. Others typically arise meiotically, and the zygote was trisomic ab initio. That is to say, meiotic or mitotic origins of the trisomy are substantially chromosome specific. For example, trisomy 8 CPM is characteristically the consequence of a mitotic event, while in contrast, almost all cases of CPM for trisomy 16 have arisen at maternal meiosis I. From a meiotic origin, “correction” may generate a 46,N karyotype in the fetus, but there is a risk for this to be associated with uniparental disomy. Thus, of the trisomy 16 CPM cases, about half displayed UPD(16) in the fetus. A meiotic origin of the CPM typically implies a more guarded prognosis than if the error had arisen somatically. Trisomy 2 at CVS is an example of a mosaicism that conveys quite different implications according the meiotic or mitotic mechanism of its generation (see later). FAL SE- N EG ATIV E RESU LTS F ROM CHOR IO N IC V ILLUS SA M PLI N G
False-negative results are very rare, and more so since many laboratories no longer use direct or short-term CVS culture. False negatives are presumed to have arisen due to an early postzygotic event, such that a normal cell line is generated in the extra-embryonic tissue from a basically abnormal conceptus; or, an abnormal cell line can arise from a normal conception, and this cell line then contributing to formation of the embryo (this latter scenario documented especially in the acrocentric isochromosome; Riegel et al., 2006). The largest formal series to address this question is due to van den Berg et al. (2006). These workers reviewed nearly 2500 prenatal diagnoses from their own service and comprehensively assessed the literature. In their own material, they had no false negatives. From the literature, most false negatives have been seen in the setting of a normal shortterm culture, and then either an abnormal long-term result,5 or, if no further testing done, an abnormal pregnancy outcome. This highlights a relative instability of the cytotrophoblast karyotype, with a tendency, as the most usual scenario in this context, to lose the additional chromosome from an initially trisomic conception. From long-term CVS culture, true negatives numbered only in single figures, and
several of these were likely due to maternal cell contamination. Thus, practically all of the time, a normal long-term CVS result means that the baby will be chromosomally normal. Fluorescence in situ hybridization (FISH), applied to direct uncultured CVS, may be chosen to enable a more timely diagnosis (a faster “turnaround time,” in the laboratory jargon), and particularly in the circumstance of an ultrasound anomaly having been seen. This can target the common aneuploidies, which account for ~65% of all chromosome abnormalities. In one large series (Feldman et al., 2000), 115 direct CVS were analyzed by interphase FISH, from pregnancies in which 100 had a minor fetal anomaly by ultrasound, and 15 had a major anomaly. All of the FISH results were confirmed by routine cytogenetics, with no false positives or false negatives as compared to the results after culturing. Although the authors did not separate the chromosome abnormalities found in CVS versus amniotic fluid, overall, they found aneuploidies by FISH in 10.6% of samples, with another 3.8% of cases having chromosome abnormalities by analysis of cultured cells that had shown a normal FISH result. Thus, the common aneuploidies are highly likely to be identified by uncultured, interphase FISH, but when the result is normal, routine karyotyping (or microarray) is still necessary to detect other abnormalities. LE VEL I I I M OSA I CI SM
Level III mosaicism in CVS raises an immediate concern. Management at this point (which will usually be around 12–13 weeks) is aimed at demonstrating, as much as possible, fetal normality; or, if it so transpires, confirming a true fetal mosaicism. Amniocentesis with rapid FISH analysis of a large number of cells, along with detailed ultrasonographic assessment of fetal morphology, is usually the next plan of action; or, in those clinics having access, a microarray analysis might be performed on the uncultured or cultured amniocytes. In fact, the majority of cases will return normal results after this additional workup, since the mosaicism is likely confined to the placenta. A large amount of data on level III mosaicism for autosomal trisomy was gathered by the European
5 Or an abnormal result from a simultaneous cord blood or amniocentesis, typically done in the context of abnormal fetal ultrasonography.
450
•
P R E N ATA L D I A G N O S I S
collaborative research group on mosaicism in CVS (EUCROMIC) (Hahnemann and Vejerslev, 1997), comprising information on just over 92,000 CVS procedures from 79 laboratories during 1986–1994. Mosaicism (or nonmosaic fetoplacental discrepancy) was seen in 650 (1.5%) cases. Of these, 192 were followed up in detail, with karyotyping of fetal fibroblasts, fetal blood, amniocytes, or neonatal tissues. Most, 84% of the 192, represented CPM. The abnormal cell line was present in either trophoblast (type I CPM; in 50%), villus mesenchyme (type II CPM; in 30%), or both (type III CPM; in 20%). A similar proportion was forthcoming from another large review, that of Phillips et al. (1996), comprising 469 cases of placental mosaicism identified at CVS in 13 separate studies, in only 50 (11%) of which was fetal mosaicism actually demonstrated. A greater risk applied when the abnormality had been detected on villus culture, and when the chromosome concerned was a marker, or one of those involved in the common trisomies. These authors emphasized the value and validity of follow-up amniocentesis. D I FFE REN T TRIS O MI ES
Certain CVS trisomies are more or less likely to reflect the same trisomy in the fetus, and the pattern and distribution of the cell lines are also indicative, as set out in Table 27–3. Trisomy 21 mosaicism on CVS is the most likely to represent a true fetal trisomy 21, whether in the nonmosaic or mosaic state. A risk applies also with trisomies 8, 9, 12, 13, 15, 18, and 20. On the other hand, CPM or fetoplacental discrepancy for trisomies 2, 3, 5, 7, 10, 11, 14, 16, 17, and 22 was never, in the EUCROMIC series, confirmed at fetal or postnatal studies. In some trisomies, a true fetal mosaicism may exist, but at such a low level that there might be no discernible effect upon the phenotype. Klein et al. (1994) reported such a case, a child born of a pregnancy in which trisomy 8 was observed in 81% of CVS cultured cells, 0% of amniotic fluid cells, and in 60% of a placental biopsy at delivery: the child had 4% and 1% mosaicism in blood at 2 and 7 months of age and 0% on a skin fibroblast study, and was normal in appearance, growth, and developmental progress at age 30 months. Of course, any fetal morphologic defect shown on ultrasonography would indicate the very substantial probability of a major degree of true fetal mosaicism, and in that case the choice of termination is appropriately offered.
Table 27–3. Outcomes in 11 Cases of Detection at Amniocentesis of a Supernumerary Marker Chromosome That Turned out to Be of X or Y Chromosomal Origin KARYOTYPE
OUTCOME
45,X/46,X,r(X) 46,X,mar(X) 45,X/46,X,der(X) 45,X/46,X,mar(Y) 45,X/46,X,der(Y) 45,X/46,X,idic(Y) 45,X/46,X,psudic(Y) 45,X/46,X,i(Yp)
t.o.p. Normal liveborn t.o.p. (normal male) Normal male liveborn Normal male at 1 year t.o.p. Normal male liveborn Male, hypospadias, testicular defect* Normal male t.o.p. (normal male) Normal male liveborn
45,X/46,X,i(Yp) 45,X/46,X,i(Yp) 45,X/46,X,r(Y)
Note: Infertility would be probable in at least some of the apparently normal males. *Absence of germinal cells of seminiferous tubules. t.o.p., termination of pregnancy Source: Schwartz et al. (1997).
RA ND O M N ESS O F SA M PL I N G
The vagaries of sampling may influence the interpretation, as the following examples show. We followed to term a woman in whom first-trimester CVS had shown trisomy 7 mosaicism with 47,XY,+7 in three out of eight clones; and yet three out of four placental samples (one from each quadrant), and peripheral blood from the (normal) baby, karyotyped 46,XY. Just one placental sample, which was not histologically distinguishable from the others, was 47,+7 (Watt et al., 1991). Presumably, the CVS sampling catheter had traversed this unrepresentative region of the placenta, and most of the sample that was eventually analyzed came from here. Similarly, in a case of i(5p) diagnosis at CVS (see later section on “Autosomal Isochromosome”), following the birth of the (normal) baby, we identified a region of placental mosaicism (Clement Wilson et al., 2002). De Pater et al. (1997) did a CVS in a pregnancy of 37 weeks gestation in which severe growth retardation and a heart defect had been identified, and this showed nonmosaic trisomy 22. However, from a simultaneous amniocentesis, only two out of ten clones were 47,XX,+22, the other eight being Chromosome Abnormalities
•
451
normal; and a cord blood from the (abnormal) baby gave a nonmosaic 46,XX karyotype. Skin fibroblasts demonstratedmosaicism,47,XX,+22[7]/46,XX[25]. Of 14 placental biopsies studied by interphase FISH, only one showed trisomy 22 cells, and at a low (about 20%) percentage. Again, it may be that a small focus of trisomic tissue happened to be in the path of the CVS sampling needle, and the sample was aspirated while the needle was at this very spot. (This case is an example of “fetal-placental mosaicism,” as illustrated in Fig. 27–4.) P ROG N OS IS
The child subsequently born provides the direct evidence of a harm, or not, due to CPM. Amor et al. (2006) undertook a detailed postnatal follow-up, from ages 4 to 11 years, of 36 children from a “CPM pregnancy,” and compared their outcomes according to a number of criteria, with a control group of 195 children having had a normal chromosome result from prenatal diagnosis. The mosaicisms included trisomies 2, 7, 8, 12, 13, 17, 18, 20, X aneuploidies, markers, and one translocation. The children from the CPM pregnancies did just as well in terms of general health, development, behavior, and intrauterine growth as did the control group. Only in respect of postnatal growth was there a small difference in favor of the control group, their mean percentiles for height and weight being 64.0 and 66.4, and with the CPM children, 51.6 and 56.8 (this might have been an effect of subtly compromised placental function). These authors did note a statistically significant increase—which does not necessarily equate to biological significance—in CPM children being perceived by their mothers as “more active,” and they were suitably cautious about this observation. There have been a very few examples of presumed CPM suspected at prenatal diagnosis, a normal follow-up amniocentesis, but with the birth subsequently of a child with the same mosaicism (Stetten et al., 2004). In these, the mosaicism was, in retrospect, clearly not confined to the placenta, but was in fact a true fetal-placental mosaicism (Fig. 27–4). UNI PA R EN TA L D IS OMY
A specific concern, when CPM for a trisomy is diagnosed, relates to uniparental disomy (UPD) (Kotzot, 2008b). This is an issue in those trisomies involving an imprintable chromosome (namely, 6, 7, 11, 14, 452
•
P R E N ATA L D I A G N O S I S
15, 16, and 20). The embryo may “correct” by postzygotic loss of the additional chromosome, while the placenta remains partly or wholly trisomic. In the particular case of trisomy 15 on CVS followed by 46,N at amniocentesis, the Prader-Willi/ Angelman methylation test can be applied. UPDs of the other chromosomes seem mostly to be without phenotypic effect per se, excepting the unlikely possibility of isozygosity for a recessive gene. Where the UPD concerned is associated, or possibly associated, with a major clinical phenotype (and see Chapter 22), prenatal testing for these UPDs (paternal upd6, upd7mat, upd11p, upd14mat and pat, upd15mat and pat, upd16mat, and upd20mat) is justified. Irrespective of imprinting, there remains also the question of a small residual trisomic cell line in the fetus, potentially contributing to an abnormal phenotype. EFFECT UPON PLACENTAL FUNCTION
If a cytogenetically abnormal cell line is confined to the placenta, does this have any implication for placental function? A global statement cannot be made: some trisomies may matter, and others not, and the fraction of placenta carrying trisomic tissue is an important variable. But for several trisomies at least, a placenta that is in part trisomic apparently retains a sufficient, or nearly sufficient level of function, and mostly the (46,N) fetus is satisfactorily supported. Lestou et al. (2000) analyzed a series of 100 placentas from pregnancies producing a “viable and nonmalformed” infant, using the methodology of CGH with confirmatory FISH, and found one with CPM in only trophoblast (trisomy 13), two with CPM in the stroma (trisomies 2, 12), and two with mosaicism in both compartments (trisomies 4, 18); thus, in these cases, the CPMs were apparently harmless. However, Robinson et al. (2010) observed placental autosomal trisomy (trisomies 2, 7, and 13) in 10% of pregnancies complicated by intrauterine growth retardation (and in some of these there was also the maternal complication of preeclampsia), but in none of 84 placentae from uncomplicated pregnancies. The more commonly observed CPMs at prenatal diagnosis involve chromosomes 2, 3, 7, and 8 (which mostly arise mitotically), and chromosomes 16 and 22 (mostly of meiotic origin, and typically affecting both trophoblast and villus core placental constituent parts). It is mostly CPM of meiotic origin that is associated with a risk for pregnancy complication (Kalousek, 2000). A quite different question is
mosaicism with “placental mesenchymal dysplasia,” in which there is a normal and a uniparental cell line (p. 396).
AMNIOTIC FLUID CELL CULTURE AND MOSAICISM A mitotic error in epiblast may produce mosaicism of both embryonic and amniotic tissue. A mitotic error in extra-embryonic epithelium causes mosaicism confined to the amniotic membrane. An in vitro cell division defect causes pseudomosaicism. Separating confined placental mosaicism and pseudomosaicism from true mosaicism is critical, but by no means straightforward. The distinction is, in the first instance, based upon the number of abnormal cells seen, and whether one or more than one presumptive abnormal clone exists, according to the three levels I-III set out earlier. Level I mosaicism is seen in 2.5%–7% of amniocenteses, level II in 0.7%–1.1%, and level III in about 2 per 1000 amniotic fluids (Wilson et al., 1989). Once the laboratory studies are completed, the cytogeneticist will provide an opinion about the level of mosaicism, taking into account technical aspects of the cultures. There is generally no point, and indeed it could be counterproductive, to report level I mosaicism. The only exception would be a single cell of a clinically relevant trisomy, and if the laboratory could not perform sufficient analysis, because of limited sample, to exclude substantial mosaicism. Some level II mosaicism and all level III mosaicism do, however, require to be conveyed to the patient, carefully and clearly interpreted. LE VE L II MOSA IC ISM
Level II mosaicism reflects a true fetal chromosomal abnormality in only 1% or less of cases (Worton and Stern, 1984; Ledbetter et al., 1992; Fryburg et al., 1993; Liou et al., 1993). The nature of the “mosaic chromosome” is important. If it is one that has been recorded, in life, in the nonmosaic trisomic state, or in the mosaic state, the level of concern is higher. This includes, for example, mosaic trisomies 8, 9, 13, 14, 15, 18, and 21 and mosaic isochromosomes 5p, 9p, 12p, and 18p. Albeit true mosaicism for many of the other trisomies has been observed in the malformed fetus in a pregnancy advancing well into the third trimester or in an abnormal liveborn child, these cases are so rare that a level II amniotic fluid mosaicism is still more likely due to artifact than a
true significant fetal mosaicism. High-resolution ultrasonography provides helpful information in this context. If further cytogenetic investigation is judged desirable—and it often is—repeat amniocentesis for interphase FISH analysis is the procedure of choice, with probe choice according to the chromosome in question. A large number of cells can be analyzed, and quickly. Fetal blood sampling, formerly the mainstay, is rarely used nowadays. (It is to be noted that not all mosaicism is necessarily present in blood, and for example fetal blood sampling only infrequently, if ever, detects a mosaic cell line in trisomy 5, 12, or 20, or i(12p); Berghella et al., 1998; Chiesa et al., 1998.) Strictly speaking, no amount of investigation could ever completely exclude the possibility of a true mosaicism of the fetus, albeit the distribution of the abnormal cell line may be rather limited and possibly of unimportant phenotypic consequence. We have seen, for example, a case of level III 47,XX, +13/46,XX mosaicism at CVS, followed by the demonstration of very low-level mosaicism at amniocentesis (1/28 colonies trisomic) and fetal blood sampling (1/400 cells trisomic). At birth, a cord blood sample from the baby showed 47,XX,+13 in 1 out of 150 cells; 2/32 cells were trisomic in amnion and 1/30 and 3/30 in two placental villus biopsies (Delatycki et al., 1998). It only needed the colony from one amniocyte not to have been analyzable, or one lymphocyte to have been passed over at each blood sampling, for the true state in the baby to have gone unrecognized. The child was reviewed at age 13: she is an above average student and unremarkable on clinical examination; on analysis of 400 cells (blood and buccal cell), none showed trisomy 13 (M. B. Delatycki, personal communication, 2009). Rare similar examples exist to disquiet the counselor (Terzoli et al., 1990; Vockley et al., 1991), but a sense of perspective is to be kept: for each autosome, only the tiniest number of level II mosaicisms (zero for most chromosomes) have turned out to reflect, in fact, a recognized true mosaicism of the fetus. LE V EL I I I M OSA I CI SM
Hsu and Benn reevaluated the issues in 1999, and they have set forth useful guidelines. These are presented in detail in Table 27–1. While every autosome has now had a mention as a mosaic trisomy at prenatal or postnatal diagnosis, some are very rare, and others are of questionable significance. Some reported associations Chromosome Abnormalities
•
453
may not necessarily have been causal. Hsu and Benn propose the stringent requirement that, before embarking upon an extensive workup, there be in the literature, for the particular chromosome, “two, or more, well-documented independent reports of confirmed amniocyte mosaicism with abnormal pregnancy outcomes.” The most extensive data treating the question are published in two reports from a collaboration of a number of American and Canadian laboratories: Hsu et al. (1997) with respect to the rare trisomies and Wallerstein et al. (2000) on trisomies 13, 18, 20, and 21. We make much use of this material in the commentaries later, and every prenatal diagnosis laboratory will want to have a copy of these papers readily at hand. Ultrasonography provides useful adjunctive evidence, but apparent normality cannot be taken as a guarantee. Studies for uniparental disomy may need to be considered in the case of mosaicism for chromosomes known to be subject to imprinting. Further modifications to these guidelines can be anticipated, as new data come to hand. One should always attempt to confirm a diagnosis of mosaicism, either on multiple fetal samples following pregnancy termination, or on blood and placenta in an infant. A posttermination study that did not confirm the abnormality could cause parents great distress, and it needs deciding with them beforehand whether they wish to have the results. An unconfirmed abnormality could be misleading in a twin pregnancy in which the diagnostic sample had come from a vanishing abnormal twin, but the posttermination tissue had come from the normal co-twin (Griffiths et al., 1996). Fejgin et al. (1997) refer to the “hopeful possibility” of mosaicism as a comfort to parents, with the posttermination tissue having sampled only the normal cell line. It is true that even multiple tissue sampling cannot be taken as having ruled out mosaicism, and a diagnosis of “apparent phenotypic normality” in a fetus still leaves open that a functional brain defect could have come to pass. T WI N P R EG N AN C Y
Discordant Karyotypes. Discordant karyotypes may be observed in the setting of either dizygous (DZ) or monozygous (MZ) twinning (p. 33). Selective termination of the abnormal twin is an option, albeit one that cannot assure that the normal twin will be unharmed; because twins may share circulations, the process of termination of the affected twin may lead to exsanguination of the normal one (Lewi et al., 2006). In MZ twins in which “trisomic 454
•
P R E N ATA L D I A G N O S I S
rescue” has been the basis of one twin being karyotypically normal, a risk for UPD applies, and this would be a concern in the case of a “UPD-vulnerable” chromosome.
SPECIFIC ABNORMALITIES In this section we attempt to outline the risks for phenotypic abnormality of specific chromosomal abnormalities detected at prenatal diagnosis. Since the available data often derive from terminated pregnancies in which only major anomalies are recognized, many of these risk figures may be underestimates. For example, a trisomy 21 fetus may appear normal, to the inexpert eye, on external observation, but we naturally assume mental defect would have resulted; and the same may apply to several other chromosomal imbalances. New knowledge will continue to accumulate, and what appears here is printed on paper, not in stone. The small number of aneuploidies that may exist in the true nonmosaic state are noted first. In the mosaic list, almost every chromosome is represented, although in the CVS section we do include also a few instances of nonmosaicism.
Autosomal Trisomy, Nonmosaic Trisomies 13 and 18 (and extremely rarely 8, 9, 14, and 22) are practically the only nonmosaic autosomal trisomies besides +21 that are detected at amniocentesis. Others occur but virtually all miscarry before the usual time of amniocentesis. Chorion villus sampling (CVS), on the other hand, is done at a gestational stage when a number of trisomies destined to abort have not yet done so. T RI S O M I ES 1 3 A N D 1 8
There is a high likelihood of spontaneous abortion after amniocentesis, and presumably it is somewhat higher if detection is by CVS. Earlier figures due to Hook (1983) are 43% for trisomy 13, and 68% for +trisomy 18; more recent data from Won et al. (2005) indicate a rate of fetal death in utero following amniocentesis-proven trisomy 18 of 32%, while Yamanaka et al. (2006) arrived at a figure of 27%. Data for survival of a liveborn child are due to Vendola et al. (2010), as set out in Table 27–2. But the outlook for a liveborn child is so bleak, with inevitable profound mental deficiency, barely a vestige of social response in those few who survive
beyond early infancy, and typically a requirement for full nursing care, that termination is sought by the majority of couples. Those who decide to maintain the pregnancy should know of the high perinatal and early infant mortality, the high likelihood of congenital malformation, and the rarity (but not impossibility) of survival beyond infancy (Brewer et al., 2002; Iliopoulos et al., 2006). Many would regard life-sustaining emergency surgery to the newborn as inappropriate (Bos et al., 1992); Carey (2001) emphasizes the need to bring the parents fully into the making of any decisions. An exceptional case exists in Chen et al. (2004e), in which nonmosaic translocation (or isochromosome) trisomy 13 was identified at CVS, but with only minor ultrasonographic findings, and mosaicism shown in subsequent amniocentesis (77%) and fetal blood sampling (14%). The child in due course had a major (but correctable) heart defect, some minor anomalies, and growth and development were judged normal at 8 months of age. Postnatal karyotyping of the placenta (46,XX,der(13;13)/46,XX/45,XX,-13), and postsurgical karyotyping of tissues from the child (46,XX), suggested that the initial chromosome constitution had been trisomy 13, but then with correction in a lineage which substantially contributed to the embryo. TRI SO MY 2 1
We expect most readers will have an expert appreciation of the predicted Down syndrome (DS) phenotype, but we do recommend Hunter’s (2001) review as a full and balanced account. Marteau et al. (1994) appraised the views of obstetricians, geneticists, and genetic nurses to the prenatal diagnosis of DS and recorded some striking differences. The respective proportions who would counsel nondirectively (see definitions earlier) were 32%, 57%, and 94%, and the respective proportions counseling directively in favor of termination were 62%, 40%, and 7%. About 6% of obstetricians would counsel directively in favor of continuing the pregnancy, but practically no geneticists or genetic nurses would. Having received a positive 47,+21 result, what personal factors influence the parental decision? A 7½ year study, over 1989–1997, reports the views of 145 women in Michigan (Kramer et al., 1998). Most (87%) elected to terminate the pregnancy. The deci-
sion did not differ according to parity, race, religion, nor, perhaps surprisingly, with the presence or absence of ultrasonographic abnormality. Older mothers, those who had already had children, and those whose prenatal procedure was done at an earlier gestation, were more likely to choose termination. A point to be aware of is that, with modern management, the survival of DS individuals approaches that of the general population (95% surviving at 1 year, according to recent data from Texas; Vendola et al., 2010, and see Table 27–2), but comorbidities become prevalent with age, raising questions of practicalities of care as the parents themselves age (Glasson et al., 2002). On the other hand, if fetal ultrasonography shows a heart malformation and/or growth retardation, fetal death in utero or postnatal death is probable (Wessels et al., 2003). In the study of Won et al. (2005), the rate of fetal death in utero after an amniocentesis diagnosis of trisomy 21, in 392 women who decided to continue the pregnancy, was 10%. OT H ER AUTOS O M A L T R I S O MY, A N D I N PA RT I CUL A R T R I S O M I ES 9 , 10, 2 0 , A N D 2 2
Never (almost) do other nonmosaic true fetal trisomies survive through to a stage of extrauterine viability. Schinzel (2001) catalogs no more than about two dozen each of trisomy 9 and trisomy 22 and barely one or two of possible trisomies 7, 8, and 14, with survival through to the third trimester. Miscarriage is nigh on inevitable, usually within the 8 to 14 week gestation range. An example is trisomy 10, of which very rare examples as nonmosaics at prenatal diagnosis are known, but survival to term is seen only in mosaic forms, and these infants very abnormal (Hahnemann et al., 2005). If natural abortion has not already occurred by the time the chromosomal result is received, and if there is supportive evidence otherwise, such as ultrasonographic defect, for there being a true fetal involvement, termination is appropriately offered. Schwendemann et al. (2009) reviewed the sonographic findings of fetuses with nonmosaic trisomy 9; heart defects and central nervous system malformations were the most frequent anomalies seen. Concerning nonmosaic trisomy 20, Stein et al. (2008) record five cases at prenatal diagnosis, the indication in each being the discovery of an anatomical abnormality on ultrasound, with early deaths in all except their own case, a child who in fact turned out to be mosaic on analysis of postnatal Chromosome Abnormalities
•
455
tissues. Of all the other nonmosaic trisomies, it is only with trisomy 22 that there might be, very rarely, the possibility of a term pregnancy, and in some, limited postnatal survival (Tinkle et al., 2003; Mokate et al., 2006; Barseghyan et al., 2009).
Autosomal Trisomy, Mosaic6 D E T EC TIO N AT C H ORI ON I C V I LLU S CU LTUR E (IN S O ME CA SES F OLLOWE D BY A N O R MA L K A RYOTY PE AT AM NI O CEN TES IS )
The substantial majority of mosaic trisomies for a single autosome are followed by a normal result at amniocentesis and at karyotyping of the child (or of the aborted fetus). In the EUCROMIC study, there were 192 gestations with mosaic or nonmosaic fetoplacental discrepancy for an autosomal trisomy, and in 84% CPM was confirmed. For mosaic trisomy 8, 9, 12, 15, and 20, only a single case of each was subsequently identified with aneuploidy in the fetus/child, compared with two each for chromosomes 13 and 18, and as many as seven for trisomy 21 (Hahnemann and Vejerslev 1997). With respect to mosaicism for multiple (>1) autosomal trisomies, the presence or absence of a normal cell line is the key point: a fetal involvement is practically never seen if there is a normal cell line, and practically always seen if there is no normal cell line (M. D. Pertile, personal communication, 2002). The general rule that Robinson et al. (1997) advance is this: CVS mosaicism due to a preconceptual (meiotic) error conveys a significant risk for fetal trisomy/UPD, whereas a postconceptual (somatic) error is usually innocuous. Mosaic trisomies 15, 16, and 22 are mostly in the former category, for example, while trisomies 3 and 7 are typically of mitotic origin, and mosaic trisomy 2 can be either. The possibility remains for a residual effect due to (1) undetected (and presumably low-level) mosaic trisomy of the fetus; (2) uniparental disomy of the fetus; and (3) placental dysfunction as a consequence of a regional placental trisomy. The risks for these scenarios differ for different chromosomes, and we provide specific commentaries following. A rich source of information is the United Kingdom
Association of Clinical Cytogeneticists database, at http://acccvs.ncl.ac.uk, which assembles the results and findings from practically every U.K. CVS laboratory over the period 1987–2000 (Wolstenholme et al., 2006). Uniparental disomy may be a concern, when an “imprintable” chromosome is involved. Kotzot (2008b) proposes that upd14pat, and upd15pat or mat, warrant testing, when a risk exists. The case is less certain with respect to paternal UPD for 11p, upd14mat and upd16mat, given the variability of the phenotypes. Although the milder clinical pictures associated with upd6pat and upd7mat might be seen as making a lesser case for investigation, clinical management can be anticipated with upd6pat, and parents may want to understand the possible clinical outcome for upd7mat. Otherwise, uniparental isodisomy for any chromosome might rarely lead to an autosomal recessive disorder, but this is scarcely predictable in an individual case (unless there should happen to be a family history of a specific recessive condition on the chromosome of concern). Mosaic Trisomy 2 at Chorionic Villus Sampling. Two broad groups of trisomy 2 mosaicism are recognized (Robinson et al., 1997; Albrecht et al., 2001; Wolstenholme et al., 2001b). In the first, a majority (~90% of the total) are characterized by a small fraction of trisomic cells, and usually seen only in cultured mesenchymal cells. The pregnancy outcome is typically normal; in the series of Sago et al. (1997), 11/11 pregnancies had a normal outcome. It may be that these cases reflect a postzygotic generation of the trisomic lineage in a restricted region of chorionic tissue in an otherwise normal conceptus, and this small trisomic region has no discernible effect upon placental function. The second, minority group is presumed due to trisomy “correction” in a 47,+2 conceptus, from either a maternal or paternal error. The level of trisomic cells in the CVS is typically high, up to 100%, with the involvement of both trophoblast and the mesenchymal core. The placenta being substantially trisomic apparently compromises its function, and IUGR is a frequent observation, with a poor outcome (Roberts et al., 2003). One case is known of the very severe defect
6 As noted earlier, a few instances of apparent nonmosaic trisomy at CVS are also included here, on the assumption that—in the circumstance of a semblance of normal fetal development—a true fetal nonmosaic trisomy for that chromosome would in fact be improbable. We assume in these cases, rather, that this would be either “fetal mosaicism, nonmosaic placenta,” or “fetal-placental mosaicism” with the sampling needle missing the karyotypically normal tissue, each of these scenarios being demonstrated in Figure 27–4.
456
•
P R E N ATA L D I A G N O S I S
of “body stalk syndrome” associated with mosaic trisomy 2 at CVS (Smrcek et al., 2003). Mosaic Trisomy 3 at Chorionic Villus Sampling. In the EUCROMIC study, of 10 cases with trisomy 3 at either short- or long-term culture, none proved to have fetal involvement, apart from one child with a normal karyotype at amniocentesis and a very low 1/100 trisomy 3 count on blood as a newborn (Hahnemann and Vejerslev, 1997). Zaslav et al. (2004) identified a case of trisomy 3, in which the initial amniocentesis showed 47,XX,+3[8]/46,XX[27], and a repeat procedure 47,XX,+3[1]/46,XX[18]. Fetal blood was normal in 100 cells. The baby was apparently normal at birth, except for IUGR. FISH of placenta demonstrated the trisomy 3; thus, it would likely have been found by CVS, had this procedure been performed. Mosaic Trisomy 4 at Chorionic Villus Sampling. This is very rare; there were none in the EUCROMIC study. Two cases are recorded in Kuchinka et al. (2001). In one case, subsequent amniocentesis gave a 46,XX karyotype, but fetal demise occurred at 30 weeks, associated with considerable growth retardation (although no externally observable malformations). Upd(4)mat was demonstrated. It remains open whether the unfortunate outcome was the consequence of the UPD or due to placental trisomy. The second case did not proceed to amniocentesis; biparental disomy 4 was demonstrated in the child. Follow up at 1 year raised some reservation: although development was judged to be normal, growth indices were low, including a head circumference at about the 3rd centile (in other words, borderline microcephaly). To complicate the story, mother and child carried a balanced t(10;15). The case in Marion et al. (1990), and followed up several years later (Brady et al., 2005), was actually an amniocentesis diagnosis, but since postnatal studies showed trisomy 4 mosaicism in the placenta, it is not unreasonable to consider this is a potential CVS example. The child, at age 14, had a low-normal intellect, and some physical body asymmetries (of hand, ear, and breast). Blood was 46,XX; skin biopsy confirmed constitutional +4 mosaicism. In another case, Gentile et al. (2005) identified mosaic trisomy 4 by amniocentesis (22% of cells). The pregnancy presented at 22 weeks gestation with micrognathia, abnormal brain development, and spinal and cardiac defects. At termination, trisomy 4 mosaicism was confirmed in placental and fetal skin cultured cells;
the cord blood karyotype was normal. Molecular analysis excluded uniparental disomy of chromosome 4, but showed that the trisomy 4 was of maternal meiotic origin. An extraordinary example of mosaic trisomy 4 at CVS with double mosaicism for trisomies 4 and 6 at amniocentesis, 47,XY+4/47,XY,+6/46,XY, is described in Wieczorek et al. (2003). The double trisomy mosaicism was confirmed on skin (but not blood) karyotyping in the child, whose phenotype, while certainly abnormal, was less so than might have been anticipated. Mosaic Trisomy 5 at Chorionic Villus Sampling. Only three cases are recorded in the EUCROMIC study; in none was a fetal trisomy subsequently shown (Hahnemann and Vejerslev 1997). Mosaic Trisomy 6 at Chorionic Villus Sampling. Very few examples are known. A detailed case report is given in Miller et al. (2001). A young mother had a 12-week CVS because of ultrasonographic anomalies (crown-rump length at 11 week size, nuchal translucency), with 60% of cells in short-term culture and 22% of long-term cells showing 47,XX,+6. Amniocentesis was declined. An abnormal heart rate at 25 weeks led to emergency delivery, and a growth-retarded infant with numerous anomalies was born. Her blood karyotype was normal, but trisomy 6 cells were found in placenta and umbilical cord samples. Growth indices remained below the third centile. On follow-up at age 2¾ years, neurodevelopmental progress was “near normal.” Skin taken at the time of surgery showed 3% (hand) and 20% (inguinal area) mosaicism. The only two other cases on record involved mosaicism on direct preparations, followed by termination in one, and an apparently normal child subsequently born in the other. Mosaic Trisomy 7 at Chorionic Villus Sampling. This is typically a mitotically arising mosaicism. Kalousek et al. (1996) looked at 14 cases of trisomy 7 CVS mosaicism and fetoplacental discordance, the fraction of trisomy ranging from 7%–88% in eleven, and with three showing 100%. Twelve infants were judged normal, and in the eight of these tested, all proved to have biparental inheritance. Two infants were of low birth weight, and the one of these tested was the only of the series with UPD and a meiotic origin; the cultured CVS in this case was 100% trisomic. In a case we studied, mentioned also Chromosome Abnormalities
•
457
earlier, three postnatal placental samples karyotyped normal, and one with trisomy 7; the baby was normal (Watt et al., 1991). In the EUCROMIC study, of 32 cases with trisomy at either or both short- and long-term culture (including three with nonmosaic trisomy), none proved to have fetal involvement (Hahnemann and Vejerslev 1997). The conclusion is that the great majority of trisomy 7 mosaicism detected at CVS arises mitotically, does not imply a risk for UPD, is confined to the placenta, does not obviously compromise intrauterine growth, and is associated with the birth of a normal baby. Mosaic Trisomy 8 at Chorionic Villus Sampling. A well-recognized postnatal phenotype (Warkany syndrome) accompanies trisomy 8 mosaicism, which may also include an increased risk for cancer (Seghezzi et al., 1996). Fetal defects are recorded on pathology examination ( Jay et al., 1999). Typically, the mosaicism is the consequence of a postzygotic nondisjunction in an initially 46,N conceptus (Danesino et al., 1998). Van Haelst et al. (2001) reviewed their experience over the period 1986– 2000, based on 33,870 prenatal tests, among which were six cases of trisomy 8 mosaicism diagnosed at CVS. These six CVS cases, as it transpired, each reflected a confined placental mosaicism, and from the five pregnancies continuing a normal baby was born. A seventh case had been reported as 46,XY normal on short-term CVS culture, but the abnormal baby had mosaic trisomy 8; thus, a false-negative diagnosis. This circumstance calls to mind the scenario proposed by Wolstenholme (1996): true fetal mosaicism is typically associated with low levels of trisomy 8 in trophoblast cells (short-term CVS culture), high levels in extra-embryonic mesoderm (long-term CVS culture), and low levels in amniocytes and fetal blood cells. Mosaic Trisomy 9 at Chorionic Villus Sampling. Saura et al. (1995) presented seven cases of trisomy 9, five of which gave a nonmosaic result, with the outcomes being abnormal in most. In the EUCROMIC study, of nine cases with trisomy 9 at either or both short- and long-term culture (including three with nonmosaic trisomy in one or both cultures), one proved to have fetal involvement (Hahnemann and Vejerslev, 1997). This single case had nonmosaic trisomy at both short- and long-term culture. Slater et al. (2000) report a case of trisomy 9 nonmosaic at CVS, but with level II mosaicism found at amniocentesis, with only two cells 458
•
P R E N ATA L D I A G N O S I S
47,XX,+9. At fetal blood sampling, all 85 cells analyzed were 46,XX. Molecular studies revealed upd(9)mat. A blood sample from the newborn infant had the karyotype 47,XX,+9[4]/46,XX[50]; upon further review of the fetal blood, 3 out of 102 cells were trisomic 9. Minor anomalies were noted in the child, who had been followed up to age 1 year. It is probable that this phenotype reflected a minor degree of residual trisomy in the child’s soma. Mosaic Trisomy 10 at Chorionic Villus Sampling. In one case, direct culture showed trisomy 10 mosaicism, while long-term culture and amniocentesis were 46,XY, but with upd(10)mat. The child subsequently born was apparently normal ( Jones et al., 1995). Mosaic Trisomy 12 at Chorionic Villus Sampling. Hahnemann and Vejerslev (1997) and Sikkema-Raddatz et al. (1999) describe three cases, two of which involved a true fetal mosaicism. Of these latter, one fetus appeared grossly normal post termination, and one infant was abnormal. Mosaic Trisomy 13 at Chorionic Villus Sampling. A high level of trisomy 13 cells may well reflect significant mosaicism of the fetus. Ultrasonography and amniocentesis, and possibly fetal blood sampling, may clarify the picture. Mosaic trisomy 13 may present a very abnormal postnatal phenotype (Delatycki and Gardner, 1997). A difficulty arises in the case of very low-level (a per cent or so) mosaicism, in which case it is possible the child could be normal (Delatycki et al., 1998). In the EUCROMIC study, of 15 cases with trisomy 13 at either or both short- and long-term culture (including four with nonmosaic trisomy in one culture), two (14%) proved to have fetal involvement (Hahnemann and Vejerslev, 1997). In a series of 6820 CVS cases, Schuring-Blom et al. (2002) identified three cases of trisomy 13 mosaicism, of which two were false positives. Mosaic Trisomy 14 at Chorionic Villus Sampling. Only three examples of 47,+14/46,N were recorded in the EUCROMIC study, none showing fetal trisomy (Hahnemann and Vejerslev, 1997). In their case, Ralph et al. (1999) proceeded to follow-up amniocentesis, which also showed the mosaicism, and in addition maternal uniparental isodisomy 14 was demonstrated. Fetal death in utero supervened; no morphological abnormality was
identified. Other prenatal cases (or retrospectively diagnosed, on postnatal placental biopsy) with the syndrome of maternal UPD14, following “correction” of trisomy, are known (Morichon-Delvallez et al., 1994; Towner et al., 2001; Engel and Antonarakis, 2002). Growth restriction, and possibly dysmorphism and minor anomalies, may be associated. Mosaic Trisomy 15 at Chorionic Villus Sampling. In two EUCROMIC studies, cases of trisomy 15 CPM were examined, in which direct and long-term cultures had been done (European collaborative research on mosaicism in CVS [EUCROMIC] 1999; Hahnemann and Vejerslev, 1997). Few of these cases demonstrated true fetal mosaicism. Most often, the trisomy 15, mosaic or nonmosaic, was found in cytotrophoblast and villus mesenchyme, and rarely in the fetus. The authors theorize that chromosome 15 (and 16) participates more often in trisomy rescue. This would increase the potential risk for upd15, and more often than not, the trisomy 15 would be meiotic in origin. The recommendation is that amniocentesis be offered to all patients with a CVS diagnosis of mosaic or full trisomy 15, prudently to check for the possibilities of UPD, and true fetal mosaicism. Redaelli et al. (2005) propose that the CPM might of itself lead to a phenotype, from their study of a case of severe IUGR and trisomy 15 CPM. At birth, mosaic trisomy 15 (84%) was shown by FISH on placental biopsy (which one could regard as equivalent to a CVS). The child had multiple malformations, including heart, gut, and genital, and an abnormal thymus. Postnatal chromosome analyses of blood and skin fibroblasts were normal, as had been an amniocentesis; upd15 was excluded. The child died at 6 months of age. Mosaic Trisomy 16 at Chorionic Villus Sampling. Almost all CPM for trisomy 16 (which may present as mosaic or nonmosaic trisomy 16 on CVS) is due to a maternal meiosis I nondisjunction. The important follow-up investigation is an amniocentesis. If this gives a normal karyotype, CPM is very probable. IUGR with a low birth weight is common, but catch-up growth is typically observed. Malformation may be present, but usually these are minor or surgically reparable birth defects. Normal intellectual capacity is well recorded (Langlois et al., 2006; Neiswanger et al., 2006). However, a more severe phenotype may result, and ultrasonography may indicate this likelihood, the complications
including major malformation, and fetal death in utero. The degree of severity may relate to the presence or absence of fetal trisomy (which may not be revealed until post-natal tissue sampling), or, in the case of CPM, to the existence of uniparental or biparental disomy of the fetus, although this latter point is controversial (DeLozier-Blanchet, 2002; Eggermann et al., 2004; Langlois et al., 2006; Neiswanger et al., 2006). If trisomy 16 mosaicism is seen at amniocentesis, the prognosis is less favorable: see later. Mosaic Trisomy 18 at Chorionic Villus Sampling. In the EUCROMIC study, of 29 cases with trisomy 18 at either or both short- and longterm culture (including eight with nonmosaic trisomy in one or both cultures), four (14%) proved to have fetal involvement (Hahnemann and Vejerslev, 1997). Harrison et al. (1993) studied placental karyotypes from pregnancies in which trisomy 18 had been diagnosed, whether at pre- or postnatal diagnosis, and mosaicism was detected in 7 of 12, involving the cytotrophoblast. This supports the view that mosaic trisomy 18 at CVS may on occasion reflect a full trisomy of the fetus (and also leads to the conclusion that fetal survival may, in the context of this particular trisomy, be enhanced if there is a diploid placental fraction). Mosaic Trisomy 20 at Chorionic Villus Sampling. Mosaic trisomy 20 is one of the commonest mosaicisms detected at amniocentesis (see later), but observation at CVS is less frequent. In the EUCROMIC study, of 12 cases with trisomy 20 at either short-term, or at both short- and long-term culture (including four with nonmosaic trisomy in short-term culture), one (8%) proved to have fetal involvement (Hahnemann and Vejerslev, 1997). Six cases were reported by Robinson et al. (2005), two of which had compromised outcomes: developmental delay in one, and growth retardation and stillbirth in the other; follow-up amniocentesis had shown trisomy at levels of 11% and 59%, respectively. Steinberg Warren et al. (2001) described a child, followed to age 8¾, normal other than hypomelanosis of Ito, from a pregnancy with a nonmosaic trisomy 20 diagnosed at CVS; culture from a subsequent amniocentesis failed. As the pigmentary skin sign in the child indicated, he was in fact mosaic, and proven to be so on skin culture; and this mosaicism would probably have been revealed, had the amniocentesis been successful. We may presume the likely Chromosome Abnormalities
•
459
circumstance as depicted in “Fetal mosaicism, nonmosaic placenta” in Figure 27–4. Mosaic Trisomy 21 at Chorionic Villus Sampling. Chromosome 21 naturally commands special attention. In the EUCROMIC study, of 22 cases with trisomy 21 at either or both short- and long-term culture (including eight with nonmosaic trisomy in one culture), nine (40%) proved to have fetal involvement (Hahnemann and Vejerslev, 1997). Beverstock et al. (1998) report a “near false-negative” finding of mosaic trisomy 21, in which trisomic cells were observed in long-term CVS culture, and then, at follow-up amniocentesis, in only one culture. True mosaic trisomy was proven at fetal blood sampling, and tissue culture post abortion. Mosaic Trisomy 22 at Chorionic Villus Sampling. Fetal defect is typically associated, but the degree may vary considerably. Wolstenholme et al. (2001a) described their own case of nonmosaic trisomy 22 diagnosed at direct and cultured CVS, with 47,XX,+22/46,XX mosaicism subsequently shown at amniocentesis (3/60 cells +22) and fetal skin biopsy (6/170 cells +22). Fairly subtle fetal dysmorphism was noted post termination, and multiple tissue samplings showed mostly low but consistent trisomy mosaicism: 1% trisomic cells in skin, muscle, blood, kidney, 3% in lung, 5% in liver, and 21% in spinal cord. It is probable that neurological compromise would have transpired, quite likely of severe degree, had the child been born. Wolstenholme et al. reviewed 11 other cases of mosaic and nonmosaic trisomy 22, the mosaicisms mostly being of high percentages at CVS, and (in the six cases proceeding to amniocentesis) low percentages at amniocentesis. Of nine cases in which posttermination samplings were done, six showed mosaicism in at least some tissues (see also the case of De Pater et al., 1997, mentioned earlier in the section on “Level III Mosaicism”). In the three cases with 0% trisomy at fetal sampling, all had manifested severe intrauterine growth retardation. This may have been the consequence of functional insufficiency of the trisomic 22 placenta; there is also the point that occult fetal trisomy can never be excluded. Bryan et al. (2002) studied a child born of a pregnancy with a nonmosaic 47,XY,+22 karyotype having been shown at CVS. There was IUGR, but the child apparently showed postnatal catchup. He typed 46,XY on peripheral blood (with biparental disomy) and was phenotypically normal, except for hypospadias. 460
•
P R E N ATA L D I A G N O S I S
DE T ECT I O N AT A M N I OT I C F LUI D CE L L CULT UR E
Considering the three major trisomies, Hsu et al. (1992) have determined that mosaicism for chromosomes 13, 18, and 21 very frequently predicts fetal abnormality, in half or more of cases. As for rare trisomies, Hsu et al. (1997) have undertaken a wide survey, based on the experiences of a number of American and Canadian laboratories and drawing on previous reports in the literature; the reader wishing full detail will need to refer to the original document. Some mosaic trisomies are associated with a high risk for phenotypic abnormality in the fetus or term infant, with figures of >60% for mosaic trisomies 2, 16, and 22, while trisomies 7, 8, and 17 are toward the lower end of the scale (<20%). Ultrasonography has a role in the assessment: most cases in which the mosaicism involves the fetus to a substantial degree will display morphologic/growth abnormality. Nevertheless, normal ultrasonography cannot allow firm reassurance. Some mosaic states might cause structural defects too subtle to be discerned at fetal imaging, and yet be associated in the child with considerable, possibly severe functional neurological compromise. In chromosomes known to be subject to parent-of-origin imprinting, uniparental disomy needs also to be factored in to the assessment. Comments on individual trisomies follow. These are rare observations, and in the survey of Forabosco et al. (2009), the most frequent mosaic autosomal trisomies recognized at amniocentesis were, in descending order: trisomies 21 (1 in 4000 amniocenteses), 20 (1 in 5000), 13 and 18 (1 in 22,000), 9 (1 in 30,000), and, each at 1 in 90,000, trisomies 2, 6, 7, 8, 15, and 17. Mosaic Trisomy 2 at Amniocentesis. In Hsu et al.’s (1997) survey, trisomy 2 conveyed the highest risk of any of the “rare trisomic” autosomes for an abnormal outcome, namely 90%, with a variable pattern of major defects. It is probable that mosaic trisomy 2 detected at amniocentesis would be in the same group as the high-level mosaic CVS case (see earlier). A trisomic line in the fetus/child may take some diligence to find. Sago et al. (1997) reported a case in which there was level III mosaicism with trisomy 2 cells present in 27% of amniocytes (and biparental disomy). The child was severely abnormal, and while blood and skin karyotyped as 46,XY, 4% of liver cells were 47,+2.
Mosaic Trisomy 3 at Amniocentesis. Only two cases were identified in Hsu et al.’s (1997) review, in one of which the child had multiple malformations, with the mosaicism confirmed on skin fibroblast culture. The child in the other case was normal. Marked intrauterine, and subsequently postnatal growth restriction, was the prominent feature in the cases in Zaslav et al. (2004) and Sheath et al. (2010); in both, development in early infancy was judged to be normal. Mosaic Trisomy 4 at Amniocentesis. A very few cases have been reported, with normal and abnormal outcomes both, approximately proportional to the fraction of trisomic cells (see trisomy 4 in CVS earlier; Marion et al., 1990; Zaslav et al., 2000; Chen et al., 2004a; Gentile et al., 2005). A single-digit percentage of +4 cells has been associated with normality, on follow-up to 1 year of age. Likewise, normal ultrasonography is a positive pointer. Mosaic Trisomy 5 at Amniocentesis. Hsu et al. (1997) recorded five cases. In one, a high level of trisomic cells (80%) was associated nevertheless with a phenotypically and karyotypically normal infant. In two, the child was abnormal, both showing the mosaicism on postnatal study. Brown et al. (2009) identified 50% mosaicism in a pregnancy in which an ultrasonographic heart abnormality had been seen, and subsequently terminated at 21 weeks. A dysmorphic facial appearance was noted, and the cardiac defect confirmed, at fetal pathology. The trisomy was confirmed in fetal tissues (excepting blood), although at a lesser level of mosaicism that that of the amniocyte culture. Mosaic Trisomy 6 at Amniocentesis. Hsu et al. (1997) recorded three cases, each with the same low-level (6%) trisomy in amniocytes, and each with a normal outcome. Reports exist of the diagnosis following recognition of fetal defects at ultrasonography, the defects ranging from minor to severe (Wallerstein et al., 2002; Wegner et al., 2004; Destree et al., 2005). One case of fetal death in utero at 23 weeks was associated with 48% trisomy cells on fetal skin analysis (Cockwell et al., 2006). Chen et al. (2006b) report a case with low-level (3%– 10%) mosaicism, with normal fetal blood karyotype, biparental inheritance, in which the parents chose termination, and the trisomy was absent on cultured fetal tissue. They suggest low-level trisomy 6 mosaicism may be a benign finding.
Mosaic Trisomy 7 at Amniocentesis. Hsu et al. (1997) recorded eight cases, with fractions of trisomic cells ranging from 5% to 48%. Only one resulted in the birth of a phenotypically abnormal child, but low-level 47,XY,+7/46,XY mosaicism was confirmed in two phenotypically normal children on foreskin analysis. Warburton (2002) emphasizes that this relatively low-risk assessment is the appropriate one to offer, and she notes also that UPD7, while unlikely, may be worth testing for. A low-level mosaic case, taken to termination with pathology study and multiple fetal karyotyping, with entirely normal findings, led these authors (Chen et al., 2005b) to agree with the view that optimistic advice may usually be appropriate. Other cases with abnormal outcome (and in which ascertainment was necessarily biased) include the following. Mosaicism was verified postnatally on skin fibroblast analysis in the child reported in Kivirikko et al. (2002), in whom fetal blood sampling and midtrimester ultrasonography had been normal; there was facial asymmetry and mild dysmorphism along with rather impressive hypomelanosis of Ito, while mental development was “considered to be within normal limits,” although no detailed assessment had been done. The fraction of trisomic colonies in the 47,XX,+7/46,XX case of Bilimoria and Rothenberg (2003) was rather high, at 41%, and in addition uniparental heterodisomy was shown in the 46,XX line; the pregnancy had come to attention because of an increased-risk maternal serum screen. On a neonatal blood sample, all cells were 46,XX, while on the contrary all placental cells analyzed were trisomic. The child was small for dates and had some minor anomalies. These authors mention an anecdote of a trisomy 7 mosaic woman “graduating from college and getting married.” Petit et al. (2011) describe a case of intrauterine growth retardation leading to amniocentesis, which was interpreted at the time as normal. The child proved to be retarded in growth and development, and displayed hypomelanosis of Ito. Blood analysis showed 46,XY with maternal UPD 7. The skin fibroblast karyotype, however, was 47,XY,+7/46, XY; and restudy of stored amniocytes from long-term culture showed mosaic trisomy 7. These authors provide a useful review. Mosaic Trisomy 8 at Amniocentesis. Counseling is difficult, and advice must be cautious. An observation of trisomy 8 in amniocytes predicts a distinct probability, but by no means a certainty, of Chromosome Abnormalities
•
461
the clinical syndrome. It is not possible to put a good figure on the level of risk. Vice versa, a true fetal mosaicism may not necessarily be detected at amniocentesis (Wolstenholme, 1996). A finding of apparently normal morphology at fetal examination following termination in some 47,+8/46,N pregnancies might be misleading, since the physical component of the clinical syndrome is relatively minor (Hsu et al., 1997). In the series of van Haelst et al. (2001) mentioned earlier, the two cases of trisomy 8 mosaicism detected at amniocentesis both turned out to be pseudomosaicism. Mosaic Trisomy 9 at Amniocentesis. The risk is high (Saura et al., 1995; Chen et al., 2003a). Hsu et al. (1997) recorded data on 25 cases, with pregnancy termination being done in 21. Abnormality was identified in most of the 21, and mosaicism confirmed in the seven having skin fibroblast studies. In the four pregnancies continuing, one abnormal child was born, with 47,+9/46,N mosaicism on blood karyotyping, the other three pregnancies resulting in apparently normal newborns. An overall figure of 56% applies for the risk that the fetus is abnormal. This high percentage figure is not surprising, and indeed it may well be an underestimate of the risk for functional abnormality in the child (intellect not being assessable in the newborn), considering the well-recorded phenotype of mosaic trisomy 9 in older individuals. A review of surviving individuals whose parents participated in a survey is given in Bruns (2011). Mosaic Trisomy 10 at Amniocentesis. In one case listed in Daniel et al. (2004), at amniocentesis a 47,XX,+10[27]/46,XX[83] karyotype was associated with severe fetal defects, this observation being the basis of the referral for prenatal diagnosis. They were able to ascertain that the cause was a postzygotic duplication of the maternal homolog. Mosaic Trisomy 11 at Amniocentesis. Of the four reported examples, all have had a normal outcome. One child came from a pregnancy with a 26% fraction of trisomic cells, with 46,N findings on postnatal tissues, and followed through to 1 year of age. Basel-Vanagaite et al. (2006) raise the question that this mosaicism may typically be a benign finding. Mosaic Trisomy 12 at Amniocentesis. This is one of the more frequently described mosaicisms, and often implies a high risk. Hsu et al. (1997) 462
•
P R E N ATA L D I A G N O S I S
accumulated 23 cases, comprising 12 continuing pregnancies and 11 terminations. In most of those proceeding to fetal or neonatal fibroblast karyotyping, the mosaicism was subsequently confirmed, albeit that most of the fetuses appeared to be normal. It is possible, however, that some subtle physical features, and possibly unsubtle neurological deficit, might have eventuated had these “normal” fetuses been born. The clinical range in the few recorded liveborn patients with true trisomy 12 mosaicism is very variable, from lethality in the newborn period, through to an otherwise normal man with Kartagener syndrome being investigated for infertility (DeLozierBlanchet et al., 2000). Of the 12 continuing pregnancies in Hsu et al. the outcomes were abnormal in 5, and grossly normal newborns in 7. Three of these normal infants followed for 5 months to 5 years were all judged to be continuing to be normal, and Staals et al. (2003) add another 3-year-old to this list. The proportion of trisomic cells at amniocentesis apparently is not a very helpful guide in prognosis. In one case in Daniel et al. (2004), associated with fetal defect at 18-week termination, the trisomy had resulted from a postzygotic duplication of one homolog. Mosaic Trisomy 13 at Amniocentesis. The risk for abnormality is very high. A collaboration of 23 American and Canadian laboratories provided data on the outcomes of 25 prenatal diagnoses of 47,+13/46 mosaicism (Wallerstein et al., 2000). Care was taken to exclude cases in which ascertainment had been biased by abnormal ultrasonography. In 21, the pregnancies were terminated. Various abnormalities were identified in 10 of these; the range of percentages of abnormal amniocytes was very wide, 6%– 94%, average 58%. No defect was detectable in the remaining 11 aborted fetuses, although the assessment was limited to simple inspection. Four pregnancies proceeded to apparently normal livebirth; the percentages of abnormal amniocytes in these were lower, ranging 5%–13%. Mosaic Trisomy 14 at Amniocentesis. Hsu et al. (1997) recorded data on five cases. In the three choosing to continue the pregnancy to term, the infants appeared normal, and typed 46,N. In the two opting for termination, fetal abnormality was shown, in one case comprising hydrocephaly. A risk exists for UPD 14, over and above any defect due to the mosaic trisomy per se, and this should be checked (see also earlier section on “Mosaic Trisomy 14 at Chorionic Villus Sampling”).
Mosaic Trisomy 15 at Amniocentesis. Trisomy 15 is usually the consequence of a maternal meiosis I nondisjunction. Amniotic fluid mosaicism may well reflect a true mosaicism of the fetus. In Hsu et al. (1997), six of the eleven cases recorded had an abnormal outcome, the risk being greater when the trisomy level was higher (>40%). Zaslav et al. (1998) review seven cases of low-level mosaic trisomy 15 detected at prenatal diagnosis, in each the amniocentesis having been done for advanced maternal age. All seven chose to terminate, and a variety of defects were documented in most but not all. In their own case, the trisomic cell line in the initial amniocyte analysis was at a low level: 47,XX,+15[2]/ 46,XX[37]. Fetal tissues were also at low levels (lung 2%–5%, heart 8%–15%, skin 6%–10%, on metaphase and interphase analysis, respectively), but the placenta showed 100% trisomy on metaphase analysis and 95% using FISH on interphase cells. These authors also document from the literature four cases of abnormal liveborns with trisomy 15 mosaicism. There is the additional question of upd(15)mat, the considerable phenotypic consequence of which— namely, Prader-Willi syndrome—may be superadded upon that of a trisomy 15 mosaicism. Mosaic Trisomy 16 at Amniocentesis. Neiswanger et al. (2006) conducted an exhaustive literature review of trisomy 16 mosaicism diagnosed prenatally, including 36 cases from amniocentesis; and they reported their own findings in three cases in which no prior CVS had been undertaken. Of these three, all had abnormal outcomes: IUGR but with normal cognitive development as judged at 14 months; IUGR and major malformations including cardiac dextroposition; and IUGR with hypoplastic left heart, leading to neonatal death. In their literature review, the figures for complication were as follows: infant death, 33%; prematurity, 64%; IUGR, 69%; physical anomalies, 75%; and just one assessed as a normal outcome, 3% (these figures being considerably worse than for CVS diagnosis). They note that level II mosaicism, in this context, may well reflect a true fetal mosaicism. The presence of UPD appeared not to influence the rates of prematurity or infant death; however, UPD was more frequent in those pregnancies with IUGR or infants with anomalies. Yong et al. (2003) tested for UPD in a series of infants from mosaic trisomy 16 pregnancies, and the fraction with upd(16)mat, at 40%, was close enough to the one-third expectation from random loss of one chromosome; and these infants were more
severely affected than those with biparental inheritance of 16. Mosaic trisomy 16 also carries an associated risk for the mother of preeclampsia (Yong et al., 2006). Thus, the earlier opinion of Hsu et al. (1998) is supported: “mosaic trisomy 16 detected through amniocentesis is not a benign finding but associated with a high risk of abnormal outcome, most commonly intrauterine growth retardation, congenital heart defect, developmental delay, and minor anomalies.” Rieubland et al. (2009) diagnosed two cases postnatally, noting a considerable phenotypic difference between the two, one normally grown and developing at age 11 months, but with a severe hypospadias, the other with IUGR, body asymmetry, numerous physical anomalies, and dying at 7 months; yet further illustrating the challenge in offering advice at prenatal detection. Notwithstanding, we have seen an eventual normal outcome, the child assessed at 2½ years of age, albeit delivery by cesarean section at 36 weeks had been necessitated due to fetal distress with IUGR. Trisomy 16 had been detected at high level on CVS and at amniocentesis, and low-level (8%) postnatally on buccal mucosal cells (Coman et al., 2010). Mosaic Trisomy 17 at Amniocentesis. This trisomy vexes, with normal and abnormal outcomes equally observed. Hsu et al. (1997) comment that the diagnosis “should not be taken lightly,” mirrored by Utermann et al. (2006) who state that “the clinical significance remains uncertain.” The longest follow-up is reported in Witters and Fryns (2008), a child at age 36 months, who was significantly delayed, with a developmental age of 26 months. And yet a number of normal outcomes are on record, as Abrams et al. (2005) document in their own case, with the child reportedly normal as a 2-year-old, and as they note similarly in a handful of other cases from the literature. They advise that an optimistic view is warranted, if the ultrasonography is normal. The cerebellum should be targeted, since malformation may be a feature of trisomy 17. Mosaic Trisomy 18 at Amniocentesis. The risk is very high. In the collaboration of Wallerstein et al. (2000), 31 prenatal diagnoses of trisomy 18 mosaicism were available for review. In just over half of these, the abortuses (induced termination or natural abortion) were abnormal. In 11, no defects were discerned at fetal examination. Just three pregnancies came to live birth, and these babies were apparently normal. The percentages of trisomic amniocytes in Chromosome Abnormalities
•
463
these three cases ranged from 2% to 20% (mean 9%), compared with 2%–95% (mean 37%) in those with abnormal outcome. A very rare, but recurrent abnormality is 45,X/47,XX,+18 mosaicism, in which the phenotype can vary from fairly mild to severe (Schluth-Bolard et al., 2009; Tyler et al., 2009). Mosaic Trisomy 19 at Amniocentesis. A single case is recorded in Hsu et al. (1997), and in which there was a normal outcome at live birth. Mosaic Trisomy 20 at Amniocentesis. This is one of the most commonly observed mosaic aneuploidies. Trisomy 20 may exist in three forms: as confined placental mosaicism, as placental-fetal mosaicism with an apparently normal phenotype in the child that is subsequently born, or as a fetal mosaicism with phenotypic consequence (Hsu et al., 1991). There may be no dysmorphic features, or only some “soft” signs, or rarely an unambiguous facial dysmorphism; a characteristic, if subtle syndrome is proposed (Willis et al., 2008). In certain fetal regions in which the trisomy may exist, in particular kidney and gut, the imbalance apparently has no discernible untoward effect, and in fact aneuploid cells may be cultured from urinary sediment. (Recognizing that amniotic fluid has a substantial contribution from fetal urine production, presumably some of the “amniotic fluid cells” from which the diagnosis of trisomy 20 had been made may have actually had origin from the fetal urinary tract.) In the collaboration of Wallerstein et al. (2000) comprising 152 diagnoses, ten (7%) were recorded with an abnormal outcome (six liveborns, four abortuses). There was correlation with the level of mosaicism: abnormality was observed in 20% of infants where there had been >50% trisomic cells at amniocentesis, and in 5% of those with <50%. Baty et al. (2001) reviewed 17 cases in which follow-up of the children extended beyond 1 year, of whom 12 (71%) had developed normally. The remaining five had various degrees of speech and motor delay. A more optimistic interpretation comes from James et al. (2002), who tracked down all cases diagnosed at amniocentesis in New Zealand 1991–2001, numbering 13, with follow-up well into childhood for 9 of these (the longest to age 10). The range of the trisomic fraction of amniocytes was 8%–50%. All were essentially normal, except for one child who had minor anomalies at birth, resolving by 6 months of age, and deformation due to breech delivery may 464
•
P R E N ATA L D I A G N O S I S
have been the cause, although weight was below the 3rd centile; and in the only case in which termination was chosen, rather subtle (indeed borderline) external fetal anomalies were noted, and cultured tissue showed low-level (skin 2%, kidney 7%) trisomy mosaicism. Baty et al. (2001) followed up two cases with higher fractions of trisomic cells at amniocentesis, 83% and 57% in one, and of 90% in the other, and the children, at ages 9 and 8 years, respectively, were of normal intelligence and of essentially normal morphological appearance. Each did, however, display quite prominent hypomelanosis of Ito, presumably reflecting a fairly widespread distribution of a trisomic 20 lineage, at least in skin. Nevertheless, reservation must remain. Reish et al. (1998) offer the sobering example of a 15-month-old child with considerably delayed gross and fine motor skills and poor language acquisition, who had 54% trisomic cells from a skin biopsy (a normal karyotype on peripheral blood). In the pregnancy, amniocentesis had shown a 45% mosaicism, fetal ultrasonography was normal, and the parents had been “cautiously counseled.” Likewise, Wallerstein et al. (2005) report a child who had seemed normal at birth, and 46,XX on blood, but who went on to manifest a “pervasive developmental disorder.” Trisomy 20 had been present in only 4/63 colonies at amniocentesis; trisomy was further documented in urinary sediment at age 4 years. They comment that “optimism regarding developmental outcome should be tempered with some caution.” Bianca et al. (2008) summarize the issues and advise along these lines: a second CVS or amniocentesis would add little value; fetal blood sampling is not useful, and neither is UPD analysis; the level of mosaicism does not predict outcome (this agrees with the views of some, and contradicts others, as noted earlier); and some reassurance may be gained from normal ultrasonography. Mosaic Trisomy 21 at Amniocentesis. The risk for Down syndrome is very high. The collaborative study of Wallerstein et al. (2000) accumulated 96 cases for review. Half had an observably abnormal outcome, with confirmatory cytogenetic study performed in a minority. Most of these were fetuses post termination with various abnormalities; six were liveborns, five of these having a clinical diagnosis of DS, and one an isolated heart defect. An apparently normal appearance (assessment limited to inspection in 39, autopsy in 2) was recorded in 41 aborted fetuses. Among these, 20 were submitted to
further cytogenetic analysis (repeat amniocentesis, fetal tissue, fetal blood, placenta), with 8 showing 8%–90% trisomic cells, and 12 with 0%. Seven liveborns were normal, two being followed up beyond the newborn period; none had confirmatory karyotyping. The mean amniotic fluid proportion of trisomic cells was 17%, range 6%–31%, in these normal children. This compares with a mean of 35% in those with a demonstrably abnormal outcome. But even in the group with the lowest level of amniotic fluid trisomy, 3%–10%, half had an abnormal outcome. From the whole material, a risk for phenotypic abnormality of 50% should be seen as a minimum estimate, since subtler defects at fetal or neonatal assessment would have escaped notice, and a potential compromise of intellectual function of course was not assessable. There is a maternal age association with this mosaicism (Forabosco et al., 2009). Mosaic Trisomy 22 at Amniocentesis. Hsu et al. (1997) determined a very high risk for abnormality for 47,+22/46, with 7 out of 11 outcomes being abnormal. Berghella et al. (1998) described a normal fetal blood result following trisomy 22 mosaicism diagnosis at amniocentesis, but fetal skin biopsy showed 47,+22/46, and structural abnormalities were subsequently identified in the aborted fetus. Four cases are noted in the review of Wolstenholme et al. (2001a), these all having followed an initial detection at CVS. Three out of the four showed some degree of normal/trisomy mosaicism at fetal samplings post termination. Leclercq et al. (2010) record a normal phenotypic outcome in a single case, followed up to age 4 years, albeit that the child showed the mosaicism on skin, in 6% of cells. Three other cases were abnormal at autopsy study (two following fetal death in utero, and one a medical termination). Mosaic Partial Trisomy at Amniocentesis. It is not feasible to list here recorded cases, and each must be judged on its merits. One specific example is worth noting, in that it may represent simply cultural artifact associated with a fragile site. This is mosaicism for a del(10)(q23). Zaslav et al. (2002) document a case of 46,XY,del(10)(q23)[9]/46,XY[45] detected at amniocentesis. The phenotypically normal child had the del(10q) in only 3/100 blood cells, this culture having been stressed by growth in a low-folate medium. We are aware of a handful of essentially similar case, all involving 10q23, and none resulting in a documented abnormal child.
The biology here is uncertain, as amniotic fluid is normally cultured under conditions that suppress fragile site expression. Indeed it is not clear that the known fragile site FRA10A at 10q23 (p. 261) is actually involved.
Polyploidy Triploidy. Close to 100% of the time, triploidy aborts spontaneously, but in some cases not until the pregnancy is well advanced. This being so, the offer of termination is appropriate when triploidy is diagnosed. Cassidy et al. (1977) described the emotional turmoil suffered by the family when a triploid infant, predicted to die immediately, survived for the extraordinary period of 5 months. Sarno et al. (1993) reported a unique case of complete placental/fetal discordance with triploidy on CVS and a normal diploid karyotype on amniocentesis and fetal blood sampling, with the birth of a normal baby; such a possibility warrants consideration where triploidy on CVS accompanies an ultrasonographically normal fetus. Nonmosaic triploidy typically shows ultrasonographic anomalies, and according to the diandric (partial mole) or digynic (asymmetric IUGR) nature of the imbalance (p. 287). True triploidy mosaicism is very rare (p. 289). Wegner et al. (2009) report a prenatal diagnosis, the pregnancy ending in fetal death in utero at 25 weeks, with the remarkable mixed-gender karyotype of 46,XX/69,XXY. Numerous abnormalities were revealed at anatomical pathological examination. They were able to demonstrate that the initial conception had been dispermic (one X and one Y-bearing sperm), and that the 69,XXY lineage had arisen by the delayed incorporation of the Y-bearing male pronucleus into a cell with a 46,XX nucleus; they preferred the expression “mixoploidy” to describe this scenario. A very rare case is “hypotriploidy” with 68 chromosomes. One case of 68,XX hypotriploidy was diagnosed prenatally following an ultrasound picture which was similar to that of classic digynic triploidy (Pasquini et al., 2010). Tetraploidy. Tetraploidy seen at prenatal diagnosis, in the context of normal ultrasonography, is usually an in vitro cultural artifact, or possibly a vestige from the blastocystic stage of normally occurring trophoblastic tetraploidy (Krieg et al., 2009). True tetraploidy is very rare, and Teyssier et al. (1997) record only ten cases, two of which had Chromosome Abnormalities
•
465
been discovered at amniocentesis. Ultrasonographic demonstration of growth retardation and enlarged cerebral ventricles may be typical, but rather nonspecific signs. While tetraploid/diploid mosaicism is almost always a cultural artifact, Edwards et al. (1994), having observed true normal/tetraploid mosaicism in two severely retarded individuals, nevertheless caution that a tetraploid cell line is not absolutely certain to be an innocuous finding. In a single such case at prenatal diagnosis, Meiner et al. (1998) showed 92,XXYY/46,XY mosaicism on fetal blood sampling following the diagnosis of nonmosaic 92,XXYY at amniocentesis, in the setting of growth retardation discovered at ultrasonography and confirmed at subsequent fetal pathology study.
Structural Rearrangement Structural rearrangements are seen in about 1 in 1000 cytogenetic prenatal diagnoses (Warburton, 1991). It is typically a matter of urgency to do parental chromosome studies, in order to distinguish between a familial or a de novo rearrangement in the fetus. If one parent is discovered to have the same apparently balanced autosomal rearrangement identified at prenatal diagnosis, and in the context of normal ultrasonographic anatomy, there is no firm evidence for an increased risk of fetal abnormality, and many would counsel to the effect of no discernibly increased risk. Sex chromosome rearrangements require separate attention. DE NOVO “APPARENTLY BALANCED” STRUC TUR AL REARRA NGE M E N T
A major difficulty is posed by the de novo rearrangement that, at the level of classical cytogenetic analysis, is “apparently balanced,” and when the interpretation at ultrasonography is normal. But even with the highest resolution banding, a submicroscopic abnormality (deletion or duplication, or gene disruption) may still be present. It may be that the occurrence of breakpoint(s) within a G-negative band, these being more gene dense, predicts a greater risk for abnormality (Fantes et al., 2008). Microarray analysis can, in these cases, be revealing, because small deletions or duplications at the breakpoint may be identified; however, a true disruption of a gene, with no net gain or loss of DNA, would not be detected (De Gregori et al., 2007; Baptista et al., 2008). Nevertheless, we should emphasize the observation that most pregnancies with prenatal 466
•
P R E N ATA L D I A G N O S I S
diagnosis of a de novo inversion or simple reciprocal translocation go on to produce a normal baby. Presumably, these normal cases reflect breakpoints in DNA that does not code for a gene or for a control element (or if a gene is disrupted, its haplo-state is sufficient), and in which there is no concomitant microdeletion. Of course, abnormal ultrasonography dictates a different perspective. Thus, for example, when Price et al. (2005) identified growth and anatomical abnormalities suggestive of Cornelia de Lange syndrome (CdLS), the subsequent finding of a presumed de novo translocation (father not available for testing) 46,XX,t(3;5)(q21;p13) enabled a clear interpretation, the CdLS gene being located at 5p13, and presumably disrupted by the rearrangement. On postnatal observation, one can be wise after the event. If a child with a particular phenotype has a rearrangement involving a breakpoint known to be in the region of a mendelian locus, or of other recorded rearrangements producing the similar phenotype, the conclusion could reasonably be drawn that the cytogenetic abnormality was the cause of that abnormal phenotype. For example, a child with a de novo inv(7)(p22q21.3) having a particular split hand/foot malformation would invite the inference of a causal link, given the similarity of the limb defect with other 7q21.3-22 rearrangements (Cobben et al., 1995). Sophisticated tools of the molecular cytogeneticist may reveal a hidden defect, such as an apparently balanced de novo 14q paracentric inversion in which Jiang et al. (2008) could actually show very small deletions at both breakpoints; these authors list the genes within the deleted segments and speculate about their possible contributions to the abnormal phenotype of the child in whom it was identified (and see also Chapter 9). In a normal person, on the other hand, an apparently balanced rearrangement we may take to be truly balanced. Caution should be exercised in the interpretation of apparently balanced translocations in which microarray testing detects an imbalance, and Gajecka et al. (2008a) provide several examples of gains and losses at the breakpoints in apparently balanced translocations in phenotypically normal individuals. Empiric Risk Estimation. Warburton (1991) conducted a review of major laboratories in the United States and Canada over a 10-year period, and collected data based on more than a third of a million procedures. We make frequent reference to this work. A de novo translocation was identified in
about 1 in 2000 amniocenteses, a Robertsonian translocation in about 1 in 9000, and an inversion in 1 in 10,000. She emphasizes that the outcome data are imperfect, given the lack of long-term follow-up, and the questionable accuracy of phenotypic assessment in terminated pregnancies. Having made that point, she does say “there was no case in which a live birth originally reported as normal was later classified as abnormal after longer follow-up. In fact, the opposite tended to occur: several cases described as having neonatal problems were later described as completely normal.” Small studies with follow-up into childhood have been undertaken (Gyejye et al., 2001), and these suggest that the figures presently offered are in the vicinity of the truth, but a clearer answer will require quite large numbers of children to be assessed. In an 8-year retrospective study from Taiwan, Peng et al. (2006) reviewed neonatal outcomes from 66 balanced rearrangements, but of which only 15 were de novo (reciprocal and Robertsonian translocations, inversions). If the anomaly rate in the inherited cases (2.0%) were to be taken as a baseline, the excess (4.7%) observed in the de novo cases (6.7% in total) could in theory be interpreted as the residual increased risk of anomalies due to the rearrangement; however, since the numbers in each category were so small, in fact little weight can be placed upon them. Given the long experience with prenatal diagnosis now accumulated, it is perhaps surprising that the data are as deficient as they are; or yet, if one considers the reality of what is involved in the logistics of long-term follow-up, perhaps not. A large collaborative exercise involving 29 Italian prenatal laboratories, covering the period 1983–2006, brought together the findings on a total of somewhat more than a quarter of a million diagnoses (amniocentesis, CVS, and fetal blood) (Giardino et al., 2009). From these, 246 de novo balanced rearrangements were identified: 177 reciprocal translocations, 45 Robertsonian translocations, 17 inversions, and 7 complex chromosome rearrangements. But follow-up data, in the 80% of cases where the pregnancy was continued, were insufficient to derive risk figures for clinical outcomes, due to logistic and legal considerations, albeit that the authors comment that “none of the newborns have been reported to display visible malformations.” We hope others who might be in a position to access similar data will not be too discouraged; one can offer a note of reassurance for the researcher (and for members of ethics committees) that most parents, in the slightly different
setting of having had the news of an ultrasound abnormality in pregnancy, are willing to respond to requests for information about how well their children subsequently did (Ramsay et al., 2009, and as we discuss on p. 20). Equally, it may be that microarray analysis will enable a clearer view, once our understanding of CNVs has settled down; the fine focus due to microarray might bypass the need for a risk estimate by directly recognizing a balanced, or an unbalanced, genome. As we have commented in Chapter 4, it is good to have good figures, in order to inform a reproductive decision. But an accurate percentage figure is, of course, only one factor in the equation. Each woman or couple will come with their own viewpoint, their own psychology, and their own mix of emotions. Wallerstein et al. (2006), in a retrospective survey of genetic counselors in North America, determined that the degree to which the news of a de novo rearrangement caused anxiety is a key factor in leading to a decision to continue the pregnancy or to terminate. The “Carrier Fetus” Who Will Become a Carrier Adult. We have discussed in the introductory chapter the issue of the genetic testing of children. In the case of prenatal diagnosis in which a de novo apparently balanced state is discovered, of course the child has already been tested, and “untesting” not a practical matter. Consider the example of the mosaic test result mentioned earlier, the wholearm translocation 46,XY,t(1;5)(p10;q10)/46,XY. Naturally, parents may want to know what reproductive implications this may have for their as-yetunborn child. In this example, the genetic risk for the child will be, as the reader can readily determine, essentially that of a likely propensity to miscarriage, should the translocation cell line involve the gonad. It is the counselor’s responsibility to communicate this sort of information in outline form to the parents, along with the advice that the child could, in the fullness of time, attend the clinic on his or her own behalf. The information must be clearly conveyed. It could be seen as a failure of the counselor’s duty of care if, in the next generation, an affected child were born, the parents being unaware of the genetic risk (Burn et al., 1983). DE NOVO BALANCED RECIPROCAL T RAN S LO CAT I O N
Simple Translocation. The starting point is that precedents are recorded for a de novo translocation Chromosome Abnormalities
•
467
having disrupted or compromised a locus, and therefore that the discovery of such a rearrangement at prenatal diagnosis could potentially herald an abnormal child. Of course, these translocations are to be taken seriously. Equally, the balanced carrier state (every one of which in the world must have been de novo at some point in the near or distant past) is very familiar, as Chapter 5 attests at length. Very many translocations are truly balanced, in terms of their functional genetic consequences. Thus, a normal child is very possible, and as the observations have shown, this is considerably the more likely outcome. In Warburton’s study, serious malformations were identified in 6% of pregnancies with a de novo simple reciprocal translocation, either at elective termination or at live birth. This is some 3% above the background risk of around 3% for malformation and/or serious functional defect that applies to all pregnancies. Thus, we may draw the inference that in about 3% of these de novo translocations the chromosomal defect was causative. It seems reasonable to assume that a slightly higher figure, perhaps another percent or so, should apply to the overall risk for not only major malformation but also important functional deficit, which might not become apparent until after babyhood. Normal ultrasonography would be somewhat, but not definitively reassuring. As mentioned earlier, microarray studies may well cast light. In a study of 14 prenatal diagnoses of de novo simple translocation, the ultrasonography being normal in 12, all proved to be balanced at the level of array-CGH (De Gregori et al., 2007). This type of study provides a springboard for interpretation when a de novo translocation is encountered at prenatal diagnosis, and the option of microarray is available.7 Whole-Arm Translocation. Very few de novo whole-arm translocations are recorded, “although the existing examples suggest an optimistic prognosis can be given” (Farrell and Fan, 1995). A whole-arm X-autosome translocation is mentioned later.
Complex Rearrangement. A de novo apparently balanced complex chromosome rearrangement (CCR) has a high risk for intellectual impairment and physical malformation, but equally, completely normal children have been born. Chen et al. (2006a) and Giardino et al. (2006) reviewed the published cases, in some of which amniocentesis had been triggered by an increased-risk maternal serum screen, or the observation of fetal anomaly on ultrasound. The outcomes were abnormal in around a half, the abnormalities ranging from developmental delay, through single or multiple malformation. Intuitively, the risk would be greater with a higher number of breakpoints, and Madan et al. (1997) provide support for this view. Microarray analysis may clarify whether a true quantitative imbalance exists; however, a CCR with a breakpoint occurring within a gene might (as with any such rearrangement) not be detected, as exemplified in the t(2;12;18) (q22.3;12q22;q21.33) reported in Engenheiro et al. (2008), in which the 2q22.3 breakpoint disrupted the ZEB2 gene, causing Mowat-Wilson syndrome. In a report of three CCRs diagnosed prenatally, all proved to be unbalanced upon array-CGH analysis (Dr Gregori et al., 2007). M OSA I CI SM F O R A D E N OVO ST R UCT UR A L R EA R R A N G EM EN T IN BALANCED STAT E
Reciprocal Translocation Mosaicism. True mosaicism for a balanced reciprocal translocation, 46,rcp/46, is very rarely recognized (Fryns and Kleczkowska, 1986; Opheim et al., 1995; Leegte et al., 1998). The great majority of this type of mosaicism seen at prenatal diagnosis is level I or II and is pseudomosaicism due to in vitro change. Some breakpoints (6p21, 13q14) are preferentially involved (Benn and Hsu, 1986). In terms of implications for fetal phenotype, it can usually be disregarded. True mosaicism for a reciprocal translocation has been reported at prenatal diagnosis, and Hsu et al. (1996) accumulated eleven examples showing one normal
7 And yet, a new technology may, in its early days, return a question rather than an answer. Rooryck et al. (2010) found an apparently balanced de novo 2;18 translocation in a child with oculo-auriculo-vertebral syndrome and proceeded to a microarray analysis. This showed that each breakpoint was in a gene desert, and no nearby plausible candidate genes that might have been influenced due to a position effect; and furthermore, a microduplication elsewhere on chromosome 18 was identified, not recorded as a known CNV, but which was paternally inherited. What, if any, responsibility these genomic alterations had, severally or separately, for the genesis of the child’s phenotype remains, for the moment, speculative. Had this analysis been done at prenatal diagnosis, the interpretation would have been fraught.
468
•
P R E N ATA L D I A G N O S I S
cell line and one with a balanced autosomal translocation. In no instance in which the pregnancy proceeded (nine of the eleven) had phenotypic abnormality been observed. Concerning a possible risk for unbalanced progeny in the next generation if the gonad were involved, each such case would need to be individually assessed; the parents would need to know to give their child access to the information in due course. Robertsonian Translocation Mosaicism. In four cases in Hsu et al. (1996) of diagnosis at amniocentesis of mosaicism for a balanced heterologous translocation, 45,rob/46, the outcome was normal in all (the mosaicism confirmed postnatally in the two infants studied). The specific translocations were 13q14q, 13q22q, and 14q21q. Whole-Arm Translocation Mosaicism. The mother reported in Wang et al. (1998) with 46,XX,t (10q;16q)/46,XX mosaicism was normal (although her child abnormal; see p. 98). We know of one case of level III mosaicism for a whole-arm translocation at amniocentesis, 46,XY,t(1;5)(p10;q10)/ 46,XY, with 30% of cells in three separate cultures showing the translocation, and confirmed on a cord blood sample at delivery (10 cells out of 50 with the translocation); on follow-up at age 4 years the child was normal and healthy (personal communication, D. Grimaldi and B. Richards, 2001). Complex Rearrangement Mosaicism. The only known example to our awareness of de novo mosaicism with a CCR and a normal cell line detected prenatally is that described in Hastings et al. (1999b), 46,XX,t(3;10)(p13;q21.1),inv(6)(p23q12)/ 46,XX, and this case was associated with fetal abnormality. Inversion Mosaicism. In four cases in Hsu et al. (1996) of diagnosis at amniocentesis of mosaicism for an inversion (pericentric or paracentric), 46,inv/46, the outcome was normal in all (all four were studied postnatally, with the mosaicism found in only one). D E NOVO X O R Y TO AU TOSOM E BALANCED TRANSLOCATION
X-Autosome Translocation. In the case of a de novo apparently balanced X-autosome translocation, there are the additional possible complications of (a) gonadal dysfunction if the breakpoint is within
one of the critical regions of the X chromosome, and (b) the unpredictability of the patterns of inactivation with the possibility of severe abnormality. On theoretical grounds, the risk may be about twice that for the simple autosomal translocation given earlier (Waters et al., 2001), although Abrams and Cotter (2004), reviewing the literature, arrived at a risk figure as high as 50% (and disregarding a possible risk for reproductive health). Nevertheless, in the case they report, a normal daughter, with follow-up to age 17 months, was born after amniocentesis (for advanced maternal age) had shown a de novo 46,X,t (X;6)(q26;q23) karyotype, with the normal X late replicating. They, and we, hope that further such cases will be reported. Hatchwell et al. (1996) provide the particular example of a severe phenotype associated with a whole-arm X-autosome translocation. On the specific issue of an Xp21 breakpoint, the question of Duchenne muscular dystrophy arises. Evans et al. (1993) actually showed normal dystrophin on a fetal muscle biopsy following detection at amniocentesis of an apparently balanced rcp(X;1) with the X breakpoint at p21, and so predicted the child would not have Duchenne/Becker muscular dystrophy; and their prediction proved to be correct. In a case of de novo 46,X,t(X;9)(p21.3;q22) diagnosed at amniocentesis, Feldman et al. (1999) showed apparent integrity of the dystrophin locus on FISH. Methylation analysis indicated preferential inactivation of the normal X. On these two observations, the couple decided to continue the pregnancy; but fetal demise occurred at 34 weeks, probably due to chorioamnionitis following premature rupture of membranes at 33 weeks. No fetal defects were seen; dystrophin staining of muscle was normal. Had microarray analysis been available, a clearer answer might have been forthcoming. Yq-Autosome Translocation. The balanced Yq-autosome reciprocal rearrangement, with a 46-chromosome count, has the gonosomal breakpoint in proximal Yq (the breakpoints usually given as q11, q11.2, or q12). Hsu (1994) reviewed 23 reports, in which the usual ascertainment was through infertility (oligospermia/azoöspermia) in the adult male, with a few being found incidentally and including one at prenatal diagnosis. Only three, including two from the early 1970s in which the detail of the rearrangement was less certain, were identified through a malformed child. It may be that such translocations should be regarded as conveying no greater risk for an abnormal intellectual phenotype than do Chromosome Abnormalities
•
469
reciprocal autosomal translocations, but acknowledging a frequent, perhaps inevitable compromise of fertility (p. 129). In the particular case of a de novo translocation with Yqh material on the short arm of an acrocentric (which is, to be precise, an unbalanced rearrangement), this is unlikely to be the basis of any phenotypic defect (p. 130). DE NOVO BALANCED ROBERTSONIAN TRANSLO C ATIO N
Heterologous Robertsonian Translocation. The great majority of cases will be disomic nonmosaic and of biparental inheritance, and a normal phenotype is to be expected. The risk for phenotypic defect over and above the baseline is due to UPD and, theoretically, to occult mosaic trisomy. Reviewing their own and others’ data, accumulating some 102 prenatal cases, Ruggeri et al. (2004) determine a risk for UPD of about 3%, based upon the observation of three affected cases (all three due to upd14mat). This figure is a little higher than earlier estimates, in which results from inherited and de novo cases had been pooled (Silverstein et al., 2002). But because the UPD cases all fell within the de novo group, it may be prudent to regard these separately (and in that case, to see the risk for the inherited form as being very low). Shaffer (2006) combined all studies on heterologous Robertsonian translocations and found that, if all chromosomal combinations are considered, the risk for UPD was 0.8%. If only those imprinted chromosomes are considered (robs that include chromosomes 14 and 15), then the risk of UPD was 0.6%. De novo cases appear to have a two-fold increased risk (~2%) as compared to maternally inherited Robertsonians (~1%) or paternally inherited (no cases identified in the surveys). Although no cases of paternal UPD were identified in the prenatal surveys, there are single case examples of paternally derived robs and UPD. Given these data, it may be warranted to check for UPD, more especially in the setting of one of the imprintable chromosomes (14 or 15) being a component of the translocation. UPD 15 can be tested at prenatal diagnosis using DNA methylation analysis at the 5′ SNRPN locus (Glenn et al., 2000); amniocytes rather than chorionic villi may be the preferable tissue to test (Silverstein et al., 2002).
8 The formally correct nomenclature is actually der(13;13)(q10;q10).
470
•
P R E N ATA L D I A G N O S I S
As for occult mosaic trisomy, this is a state that may have arisen from an initially trisomic 46,rob conception. The trisomic chromosome may then be discarded at a postzygotic “correction,” with the conceptus now cytogenetically balanced (45,rob), but with the possibility remaining of mosaicism with an occult, or very low-level trisomic cell line(45,rob/46,rob). While this cannot absolutely be excluded, the recorded experience to date indicates that this is an exceptional complication, at least at a level that might have evident clinical consequences. No such case was discovered in a de novo rob in the series of Berend et al. (2000a) (there was a single case due to a familial translocation that did have low-level (4%) trisomy 13 mosaicism, along with UPD 13). Homologous Robertsonian Translocation (or Acrocentric Long Arm Isochromosome). A chromosome comprising two long arms of the same acrocentric chromosome may be either an homologous Robertsonian translocation, or an isochromosome: for example rob(13q13q)8, or i(13q). If the formation of an homologous rob has been through the fusion of the maternal and paternal homologs, which of course must have occurred as a postfertilization event, then the rearrangement manifestly has to be a true Robertsonian translocation, and the inheritance is biparental. In that case, a phenotypically normal child is the expectation, other things being equal (Abrams et al., 2001); infertility would, however, be anticipated (see Chapter 7). All isochromosomes, and some homologous translocations, will display uniparental inheritance. The importance of uniparental disomy depends upon the chromosome involved. In Berend et al.’s (2000a) Robertsonian series, there were six identified with an homologous translocation, all de novo, and four of these had UPD, two upd(13)pat and two upd(14) pat. Barring isozygosity for a single gene mutation (see later), normal outcomes are to be expected following prenatal diagnosis of a Robertsonian translocation (isochromosome) comprising a chromosome not subject to imprinting (chromosomes 13, 21, 22). This is actually recorded for the i(13q) UPD (Berend et al., 1999). No prenatal diagnosis reports exist for i(21q) UPD or i(22q) UPD, but the postnatal state of normality in each of these is known (Engel and Antonarakis, 2002). Isodisomy for at
least part of the chromosome will exist in the i(13q) UPD, i(21q) UPD, and i(22q) UPD states, and this raises the question of a risk, not readily quantifiable but likely very small, for a mendelian autosomal recessive disorder due to isozygosity, the parent being heterozygous for the mutation in question. On the other hand, for the imprintable chromosomes 14 and 15, the risk for clinical defect is absolute following prenatal diagnosis of the rea(14) UPD and the rea(15) UPD, and the clinical syndromes of UPD14 or UPD15, maternal or paternal, would inevitably ensue (Berend et al., 2000a; McGowan et al., 2002). DE NOVO BALANCED INVERSION (PERICENTRIC OR PARACENTRIC)
(Note that a supposed “inversion” detected in this setting may actually be an unbalanced translocation). The risk from Warburton (1991) for phenotypic abnormality associated with a de novo inversion is 9.4%, which is 6%–7% over and above the background risk. The numerator is small, however, and the 95% confidence limits span 2%–25%. Since, in theory, a two-breakpoint inversion should not imply a greater risk than the two-break reciprocal translocation, the figure for this latter category as noted earlier, namely 3% (or a little above), might reasonably be seen as appropriate also for the inversion. Although if one breakpoint is in an acrocentric short arm, the risk might be that much less (Leach et al., 2005). In the special case of the X inversion, there may be gonadal insufficiency in the female (Dar et al., 1988; Dahoun et al., 1990). Pathology due to an inversion per se is rare, but well recognized; for example, Sotos syndrome consequential upon disruption of the NSD1 gene, due to a de novo inv(5)(q33.3q35.3) (Malan et al., 2008). DE NOVO BALANCED INSERTION
Only one case is recorded, to our knowledge, of a de novo apparently balanced autosomal interchromosomal insertion detected prenatally (Hashish et al., 1992). The child proved to be phenotypically normal. Van Hemel and Eussen (2000), in their review of nearly 90 families with an interchromosomal insertion, note that of the nine probands with congenital anomalies having a balanced insertion, seven were de novo and only two familial. It might reasonably be suggested that the risk for the interchromosomal insertion (three breakpoints) would
be similar or possibly a little greater than the de novo apparently balanced reciprocal translocation (two breakpoints). Recalling the 3% risk figure associated with the latter, perhaps a percent point above this is a fair figure to offer for the risk of “unspecified malformation and/or intellectual deficit.” Gonadal insufficiency may accompany the de novo intrachromosomal ins(X) (Grass et al., 1981). DE NOVO BALANCED AUTOSOMAL RI NG CH R O M OS O M E
The 46,r(A) ring chromosome is discussed in Chapter 11, and the reader is referred to specific instances listed therein. Rings that are truly balanced, reflecting a tip-to-tip telomere fusion, are nevertheless likely to cause growth retardation (or, in the case of r(20), epilepsy). Microarray analysis can reveal a very subtle deletion, which even targeted MLPA and FISH could not detect, as Manolakos et al. (2009) show in a case of ring 15 chromosome prenatal diagnosis (p. 205). Chen et al. (2006d) describe a specific example of a ring 9 detected at amniocentesis, with secondarily arising monosomic and trisomic cell lines flanking the disomic karyotype, 45,–9/46,r(9)/46,idic r(9), and in which subtelomeric deletions were demonstrated; post termination, fetal abnormality was evident. These authors report a similar story with a ring 4 (Chen et al., 2007a). Equally, array-CGH may demonstrate no apparent loss of material, as Papoulidis et al. (2010) report with a ring 21, the baby subsequently born being assessed as normal. DE NOVO UNBALANCED ST R UCT UR A L R EA R R A N G EM EN T
Unbalanced Rearrangement, Modal Number 46 or 45. Autosomal. For any de novo autosomal structural rearrangement in which cytogenetic imbalance can be demonstrated, serious phenotypic abnormality is highly likely. Often, it is not possible readily to identify the precise origin of a duplicated segment, which means that precise prediction of phenotype is not possible. Many cases, indeed most, are unlikely to be exactly the same as those in the literature or on the databases, and the counselor will need to make an informed evaluation. Ultrasonography may clarify the question if abnormalities are seen, but an apparently normal sonogram does not guarantee that the child would be normal (Al-Kouatly et al., 2002). Chromosome Abnormalities
•
471
In the mosaic state, the risk may be high if pseudomosaicism is judged to be unlikely. Hsu et al. (1992) record 34 cases with at least one cell line having an unbalanced rearrangement (thus, presumed to be a true mosaicism). In follow-up studies, phenotypic abnormality was noted in about 50% and cytogenetic confirmation obtained in 65%. Each rearrangement needs to be considered on its merits. The dilemma of deciding how best to advise couples is illustrated in Cotter et al. (1998). They describe the karyotype 46,XX,der(4)t(4;5) (q34;q12)/46,XX detected at amniocentesis, imparting, in the abnormal cell line, trisomy for most of 5q. This was confirmed on two subsequent amniocenteses, with an average overall of 17% of amniocytes abnormal, but with a 46,XX result on fetal blood sampling, and normal ultrasonography. The parents were advised that “few data were available” to determine risk; they made a decision to continue the pregnancy. In the event, the child appeared normal at birth and at 2-year follow-up; 100 cells at cord blood karyotyping were normal. In contrast, 46,XX,add(15)(p10),t(2;15)(p10;q10)/ 46,XX mosaicism detected at 30-week prenatal diagnosis (performed due to IUGR), and shown on both amniocentesis and fetal blood sampling, was associated, post termination, with fetal anomalies consistent with a partial trisomy 2p (Pipiras et al., 2004). Cotter et al. rightly call for others’ experience in similar cases to be published. If a “jumping translocation” (p. 159) leads to imbalance, fetal defect is very probable (Annable et al., 2008). X-Autosomal. Prediction with respect to the unbalanced X-autosome translocation is precarious (and see Chapter 6). Albeit the pattern of inactivation may lessen the effect, and indeed convert an invariably lethal imbalance to a survivable state, the degree to which selective inactivation may occur in fetal tissues is not knowable, and a significant defect remains very probable (Kulharya et al., 1995; Garcia-Heras et al., 1997; Orellana et al., 2001). Had the child with an unbalanced der(X)t(Xp;22q) described on p. 127 (see Fig. 6–10 in Chapter 6) been identified at amniocentesis, and with the DiGeorge critical region intact and no inactivation on the 22q segment, a prediction of typical Turner syndrome might have been reasonable. In the event, this child proved to have a significant mental handicap. Contrary examples in which a prediction of major abnormality would have been mistaken are rare. 472
•
P R E N ATA L D I A G N O S I S
Y-Autosomal. Autosomal material attached to the heterochromatin of a Y chromosome is to be seen in essentially the same light, as if it had been a translocation to an autosome (and see Chapter 6). A rare, but recurrent unbalanced karyotype seen at prenatal diagnosis is the t(Y;1)(q12;q21) translocation in mosaic state, which endows essentially a 1q trisomy in the tissue with the translocation (Scheuerle et al., 2005). The phenotype is lethal. Vice versa, if Y material is attached to an autosome, and if autosomal material is lost at that site, the autosomal monosomy of itself determines phenotypic defect (Klein et al., 2005). A somewhat different, and very rare category, is that in which a near-intact Y, missing only part of the pseudoautosomal region, combines with an acrocentric chromosome. Borie et al. (2004) describe the prenatal diagnosis of 45,X,dic(Y;22)(p11.3;p11). Had this dicentric chromosome included all the Yp material, the child might have been normal. But in fact the SHOX locus, at Yp11.3 (see Fig. 6–1 in Chapter 6), was deleted, and the otherwise normal male child had short stature. Yq;15p Variant. In the population there is a common variant whereby the heterochromatin of Yq becomes translocated to the short arm of chromosome 15; this occurs in about 1 in 2000 individuals. Occasionally, other translocations will occur between these two chromosomes, such is the case reported by Chen et al. (2007), in which the father’s karyotype was 46,X,t(Y;15)(q12;p13) and the female fetus inherited the abnormal chromosome 15. Because the derivative chromosome has deleted the repetitive 15 short arm and replaced it with Yq heterochromatin, no phenotypic effect would be expected. The authors suggest that methylation analysis for chromosome 15 should be considered, although in fact no cases of UPD 15 due to this common variant have been reported. UNBALANCED REARRANGEMENT, M O DA L N UM B ER 4 7 : S UPER N UM ER A R Y C HR O M OS O M E
A supernumerary chromosome may be of substantial size, and identifiable as to its makeup; or it may be smaller, and its origin uncertain. The latter are referred to as supernumerary marker chromosomes (SMCs), and these have also been described as marker, extra structurally abnormal chromosomes (ESACs), and accessory chromosomes (Hook and Cross, 1987a). The SMCs we mostly consider here
are the small SMCs (sSMC); Liehr (2008) defines these as structurally abnormal chromosomes that cannot be identified or characterized unambiguously by conventional banding cytogenetics alone, and which are generally equal in size or smaller than a chromosome 20 in the same metaphase spread. Some are quite harmless, and associated with phenotypic normality, and others are not: they are a very heterogeneous group. SMCs are encountered in about 1 in 1000 prenatal diagnoses, frequently in the mosaic state with a normal cell line. Upon the discovery of an SMC at prenatal diagnosis, an urgent parental chromosome analysis is required. The majority will prove to be de novo. These questions are to be asked: From which chromosome is it derived, and does it comprise euchromatin or heterochromatin? Is it a recognized type of SMC, for which precedents are recorded? Precise characterization is necessary, and this requires the use of FISH or other stains (Liehr, 2008; Yaron et al., 2003), or of microarray analysis (Ballif et al., 2006; Pietrzak et al., 2007; Gruchy et al., 2008). On FISH, about 80% are shown to derive from one of the acrocentric chromosomes, most commonly chromosome 15 or chromosome 22, and often involving only the pericentromeric region and/or the satellites (Crolla et al., 1998; Lin et al., 2006). De Novo Identifiable Supernumerary Chromosome of Substantial Size. An additional chromosome which is of sufficient size that it can be characterized on initial routine analysis as a deleted or rearranged form of a specific autosome will imply a very high risk of abnormality, approaching 100%, due to partial trisomy of that chromosome. Once a supernumerary chromosome has been identified, it is no longer referred to as an SMC; it is now described as a ring or derivative, for example, r(7) or der(22)9, or whatever may be the precise description. Familial Supernumerary Marker Chromosome. Interpretation in the case of a familial SMC is usually straightforward. If one parent is also 47,+SMC and phenotypically normal, it can be assumed no discernibly increased risk for fetal abnormality exists
(Brøndum-Nielsen and Mikkelsen, 1995). Hastings et al. (1999a) surveyed a 10-year experience in London and report six familial SMCs, which included three that were 14 or 22-derived, an idic(15), a der(6), and a mosaic der (16). The outcomes in the five proceeding to live birth were all normal at follow-up from 5 months through 5 years. When the parent has the SMC in mosaic state, prediction for the fetus is more difficult: the chromosome could be potentially harmful, but the parent might have been protected by a particular tissue distribution. The mosaic der(16) in Hastings et al. comprised centromeric chromatin: mother and child, both mosaic, were normal. Each case will need to be judged on its merits. The idic(22) presents an exception, since a parent can be normal and the child abnormal (Crolla et al., 1997). A unique case of the parental SMC being seen in double dose at prenatal diagnosis is recorded in Wolstenholme et al. (1992). At CVS, amniocentesis, and in the normal child, the karyotype was 48,XX,+mar,+mar, from a 47,XX,+mar mother. If the SMC is revealed as being a small derivative chromosome from 3:1 malsegregation, one parent being a balanced translocation carrier (Stamberg and Thomas, 1986), this is of course an entirely different story, and serious phenotypic abnormality is practically certain. Similarly, Nevado et al. (2009) warn of the need to beware of the sSMC from a carrier parent who has a cryptic deletion for the same material (see p. 158). Baldwin et al. (2008b) document the prenatal diagnosis of a small ring 4, which was then shown to be carried (in mosaic state) in the mother, and the pregnancy continued. When the child was subsequently investigated for speech delay, in fact he was shown to be duplicated for the genomic material carried in the ring, he not having inherited his mother’s “balancing” deleted chromosome 4. De Novo Supernumerary Marker Chromosome. De novo SMCs have been described for most chromosomes (Hastings et al., 1999a); two-thirds of small SMCs are acrocentric derived (Dalprà et al., 2005). The mode of ascertainment may suggest a category of risk: those in which fetal ultrasonographic
9 Such an example comes from the prenatal diagnosis and postnatal confirmation of 47,XX,+mar/46,XX, the small marker of either 14 or 22 derivation, in which further studies indicated the probable scenario to have been a 47,XX,+22 conception, but then with correction brought about by conversion of one chromosome 22 into the SMC (along with maternal UPD22), in the lineage of the embryo. The infant showed normal/advanced early development (Bartels et al., 2003).
Chromosome Abnormalities
•
473
anomaly has been detected would enter a higher risk group, as might, intuitively, those discovered through an increased-risk finding at maternal serum screening. The risk for abnormality is low in the very small derivatives of acrocentric chromosomes which stain negatively for euchromatin, and which may be satellited. If, however, a der(15) contains the segment of proximal 15q that includes the Prader-Willi/Angelman region, the risk is high (see later). Mosaicism appears not to alter the risk for abnormality. With a reasonable level of cytogenetic characterization of small SMCs and ultrasound examination, it is possible to categorize most fetuses as being either at high risk of abnormality, or at a relatively low risk (less or much less than 5%). In principle, those comprising heterochromatin convey a low risk, while a euchromatic SMC may imply a high risk. Published series of liveborn children with SMCs are mostly biased by ascertainment in favor of phenotypic abnormality. Series of prenatally diagnosed fetuses are deficient in that there is usually only a short-term follow-up of liveborn children, while pathological assessments following termination can only show major structural malformations (Warburton, 1991). Brøndum-Nielsen and Mikkelsen (1995) report a 10-year experience in Glostrup during which nine de novo SMCs were identified. In seven cases, termination of pregnancy was chosen, with some of these showing defects at pathological examination; and in the two pregnancies continuing, one infant with a minute acrocentric-derived SMC was normal at birth, while one with a ring-like 17 was “slightly retarded” at age 2 years. In the similar survey of Hastings et al. (1999a), data were presented on 31 prenatally diagnosed SMCs, of which 21 were de novo. In ten of these 21 proceeding to FISH analysis, six being mosaic, five were shown to be 15-derived and three 14 or 22-derived; the remaining two included a r(8) and a der(16). Of the six in which the pregnancies continued, only the r(8) child was physically and developmentally abnormal. The minute marker (the very small SMC, minSMC) may comprise only centromeric material, and we discuss such a prenatal case on p. 421, a minSMC apparently comprising no more than chromosome 18 centromere; the child turned out to be normal. A similar example, although the sSMC not quite so tiny, and the child subsequently born being normal, is given in Sung et al. (2009); the sSMC comprised chromosome 10 material (and 474
•
P R E N ATA L D I A G N O S I S
may actually have been a very small ring). Microarray in such cases might not be readily interpretable: that is to say, judgment about a possible pathogenicity may be uncertain. A 21-derived sSMC at amniocentesis, which might otherwise have been interpreted benignly, was seen in a different light due to an accompanying minor 47,XY,+21 cell line, leading to a conclusion that an initially trisomic 21 conception had generated a del(21q) cell line; upon posttermination, the fetal karyotype was 47,XY,+21/47,XY, +der(21)/46,XY (Stefanou and Crocker, 2004). Specific Well-Characterized De Novo Supernumerary Chromosomes. The Bisatellited Microchromosome. These tiny chromosomes can be thought of as the reciprocal product of the Robertsonian rearrangement. They are typically harmless (Romain et al., 1981; Adhvaryu et al., 1998; Dalprà et al., 2005; Gruchy et al., 2008). isodicentric 15. About half of all SMCs are an idic(15) (also referred to as pseudodicentric 15, or inverted duplication 15; and see p. 306). These are typically dicentric and bisatellited, although one of the centromeres may be suppressed. The smallest ones (smaller than chromosome 21q) appear to be harmless, but larger ones result in the “idic(15) syndrome,” characterized by mental defect and autistic features. The boundary between smaller and larger is in 15q12. The use of D15S10 or SNRPN FISH probes, which recognize sequences in 15q12-q13, enables distinction of harmless and pathogenic chromosomes (Eggermann et al., 2002). We may anticipate an increasing role for microarray analysis (Wang et al., 2004). Rare idic(15)s have been associated with UPD 15, and it may be warranted to check for this possibility (Hastings et al., 1999a). isodicentric 22. The bisatellited idic(22) typically, but not invariably, causes cat-eye syndrome. If the idic(22) lacks proximal 22q euchromatin, normality is very probable, whereas those containing euchromatin can lead to a phenotype anywhere between full cat-eye syndrome and normality (Crolla et al., 1997). Autosomal Isochromosomes. The mosaic state is usual for a supernumerary isochromosome, and thus the discovery of 47,+i/46,N is always a concern, whether at a level II or even level I mosaicism. Such a karyotype raises the prospect of an effective mosaic tetrasomy for the chromosomal arm concerned.
A 46-chromosome karyotype in which one homolog is replaced by an isochromosome typically implies a trisomy for one arm of that chromosome, and monosomy for the other. These are certainly rare observations: in an amniocentesis-based survey from Italy, based on a little under 90,000 diagnoses, the most frequent were, in order, isochromosomes of 20q, 9p, 18p, and 12p, at approximately 1 in 30,000, 45,000, 45,000, and 90,000, respectively (Forabosco et al., 2009). 47,+i(5p). Sijmons et al. (1993) assessed a dysmorphic and neurologically compromised child with a 5p isochromosome in 3/31 lymphocytes and 12/14 skin fibroblasts, and yet upon retrospective checking, only one of 217 cells from a stored shortterm CVS culture was 47,XY,+i(5p). We contrast this unfortunate experience with ours of seven cases of i(5p) mosaicism identified at CVS, six of which went on to follow-up amniocentesis (Clement Wilson et al., 2002). Three children were followed up to 2½, 3¼, and 4 years, and their normality was quite apparent. In one of these children, a circumscribed area of the placenta following delivery karyotyped 47,+i(5p), adjacent parts karyotyped 47,+i(5p)/ 46,N, and most of the placenta (and the child himself) had a normal karyotype. The CVS sampling had presumably needled this small region of confined placental i(5p) mosaicism. One pregnancy tested 100% i(5p) at CVS, and the parents chose termination; no i(5p) cells were detected from fetal skin culture. In another with a 65% load at CVS, a follow-up amniocentesis showed 45% of cells with the isochromosome, and posttermination tissues showed 15%–30%. From the foregoing, we may conclude that a CVS diagnosis with a normal follow-up amniocentesis and with normal ultrasonography suggests, but cannot confirm, a normal child. As for the primary detection of i(5p) mosaicism at amniocentesis, only three cases are recorded, with all three having an abnormal outcome (Reddy and Huang, 2003). 47,+i(8p). López-Pajares et al. (2003) review the small number of reported cases. Two examples are given of discordance between amniocentesis (normal) and postnatal blood (tetrasomy 8p), an unusual pattern for isochromosomes (but cf. the i(9p) later). A disconcerting story is told in Nucaro et al. (2006): i(8p) mosaicism was seen at long-term cultured (but not short-term) CVS, with a normal result after amniocentesis, but resulting in a child severely retarded and epileptic, and with a 5% level of the i(8p) on blood.
47,+i(9p). The clinical picture, and the subtleties of different breakpoints, are reviewed in Dhandha et al. (2002). Isochromosome mosaicism can be the basis of a false-negative test result at prenatal diagnosis. Thus, Eggermann et al. (1998) reported an abnormal baby born to a 39-year-old mother, in whom amniocentesis at 14 weeks gestation had returned a normal karyotype. On blood analysis, the child had an i(9p) in 32% of cells. From one skin biopsy, 50 cells had a normal karyotype, but on a second biopsy, 5 out of 8 cells showed the i(9p) chromosome. The particular attribute of the i(9p) is for blood, but not skin, to show the abnormality, and this may provide the explanation for its nondetection at amniocentesis. Pertile et al. (1996) support this interpretation, in their follow-up of a (nonmosaic) CVS diagnosis of idic(9)(q13). An extensive search at amniocentesis revealed a single abnormal colony, which might well otherwise have been missed. Finally, fetal blood sampling showed the idic(9) in 8% of cells. A more severe case is recorded in Tang et al. (2004), which showed the isochromosome in all amniocytes at 24 weeks and most blood and fibroblast cells from the multiply malformed infant (who died at 1 month of age). 47,+i(10p). A single case is on record, the diagnosis having been made following the recognition of fetal defects on ultrasonography (Wu et al., 2003). 46,i(13q). A de novo “Robertsonian” translocation, leading to trisomy 13, is, in the majority of cases, actually an isochromosome (Bugge et al., 2005). 47,+i(12p). The 12p isochromosome is the basis of the Pallister-Killian syndrome. The fractions of abnormal cells detected at prenatal diagnosis can vary greatly. Bernert et al. (1992) showed in one example 100% of short-term CVS cells and 10% of amniotic fluid cells having the 47,+i(12p) karyotype, whereas in Kunz et al. (2009), at CVS the isochromosome was seen only in long-term culture; in both cases, the pregnancies were terminated. Horn et al. (1995) reported a pregnancy in which CVS gave a 46,XY result on direct (17 cells) and cultured (8 cells) analysis (and 28 further cells on a retrospective study), and the abnormal newborn baby was 46,XY on a peripheral blood study (100 cells counted); at 18 months, a clinical diagnosis of Pallister-Killian syndrome was made, and the karyotype on skin fibroblast culture was 47,XY,+i(12p)/ 46,XY, with 85% of cells having the isochromosome. (Had it been an amniocentesis rather than CVS that had been done, abnormal cells would probably have been seen.) While classical karyotyping Chromosome Abnormalities
•
475
typically returns a normal result, array-CGH is able to detect subtle mosaicism in this tissue (Theisen et al., 2009). 47,+i(18p). Schinzel (2001) notes that over 75 cases of 47,+i(18p) have been recorded. Multiple physical anomalies and a moderate to severe degree of mental retardation characterize the clinical picture. Boyle et al. (2001) emphasize the plausibility of a premeiotic origin, and the caution therefore that gonadal mosaicism may exist in a parent, as they illustrate in their report of affected half-sisters. 46,i(18q). The karyotype produces a combination of monosomy 18p and trisomy 18q. Chen et al. (1998) record that many 18q isochromosomes diagnosed prenatally are associated with very severe malformation, such as holoprosencephaly and cloacal dysgenesis. Levy-Mozziconacci et al. (1996) describe a case presenting at 22 weeks gestation with abnormal ultrasonography, and although the direct CVS was 46,XX in all cells, amniocentesis and fetal blood sampling showed the isochromosome (an isodicentric, in this instance) in all cells: an example of complete CVS-amniocentesis discordance. 46,i(20q). An i(20q) identified at amniocentesis in mosaic form appears most often to be a benign finding, a rather surprising conclusion. It may be an unusual sort of mosaicism in being confined, or largely so, to amniocytes, the abnormal cell line having arisen as a postzygotic event, and its growth perhaps favored in vitro (Robinson et al., 2007). The few reported cases with fetal defect could reflect a tissue distribution which included the fetal anatomy. Goumy et al. (2005) counsel caution, and point to the advisability of careful ultrasonography, targeted in particular to the brain and vertebrae. 46,i(21q). This rearrangement is an isochromosome, not a Robertsonian translocation (Shaffer et al., 1991). The phenotype is that of Down syndrome. Gilardi et al. (2002) report a case in which the isochromosome probably arose postzygotically in an early cell destined to form the lineage of the inner cell mass and the extra-embryonic mesoderm, such that a direct CVS gave a nonmosaic 46,XX result, while long-term CVS and posttermination fetal studies showed nonmosaic 46,XX,i(21q), and a similar story comes from Brisset et al. (2003). The i(21q) can also exist in a 47-chromosome karyotype. Nagarsheth and Mootabar (1997) showed a 47,XY,+i(21q)[6]/46,XY[19] karyotype at amniocentesis; the parents elected to continue the pregnancy, and the abnormal child had only one 476
•
P R E N ATA L D I A G N O S I S
out of 120 peripheral blood lymphocytes with the i(21q), the other 119 being normal. These authors suggest that some previously reported cases of supposed i(12p) mosaicism may have been, in fact, i(21q). 47,+i(22q). A single case of an isochromosome for 22q being detected at amniocentesis is recorded in Guzé et al. (2004). The isochromosome was probably generated postzygotically, with the subsequent production of additional abnormal cell lines. The pregnancy continued to full term: the child had several defects and died on the second day of life. De Novo Supernumerary Ring Chromosomes (Autosomal). Autosomal ring chromosomes imply a high risk of phenotypic abnormality. They originate from a variety of chromosomes and contain euchromatin. Certain of these, in which only one arm of the chromosome is represented in the ring, are specifically recorded in association with phenotypic abnormality: r(1p), r(5p), r(7q), r(8q), r(9p), r(10p), r(20p), r(20q) (Anderlid et al., 2001). The r(8) with an abnormal outcome in Hastings et al. (1999a) is mentioned earlier. Uniparental disomy may complicate the picture: James et al. (1995) and Anderlid et al. (2001) report supernumerary rings, from chromosomes 6 and 9, associated with UPD 6 and UPD 9, respectively. Very small rings, that might also be categorized as “small supernumerary marker chromosomes” (sSMCs; see earlier) might not necessarily cause an abnormal phenotype: for example, two infants in Kitsiou-Tzeli et al. (2009) born following prenatal diagnosis of 46,N/47,+r(20) mosaicism were judged normal in early infancy. Chromosomal Breakage Resembling the ICF Syndrome. The chromosomes at CVS may on occasion somewhat resemble the “starburst” appearance in the ICF syndrome (p. 347, Fig. 21–4). Ehrlich et al. (2001) noted that “anecdotal observations of these types of pericentromeric chromosome 1 and 16 anomalies in normal CVS metaphases are common” and concluded that “ICF-like chromosomal abnormalities are part of the normal spectrum for CVS chromosomes and need not indicate any clinical condition,” a conclusion supported by subsequent study (Tsien et al., 2002). Presumed Normal Variants. Chen et al. (2006c) review the question of variants detected at prenatal diagnosis. They identified 16 variants of euchromatin
or heterochromatin in 21,832 amniocenteses. Eight of nine euchromatic variants were proven inherited, and seven were C-band positive. The remaining C-band positive, heterochromatic variants were all inherited from a carrier parent. Concerning the specific case of the NOR translocation, or interstitially inserted satellite, and as noted in Chapter 16, “genetic counseling should be reassuring” if this is discovered at prenatal diagnosis (Faivre et al., 1999, 2000a; Chen et al., 2004b). The Y;15 variant is noted earlier.
Sex Chromosome Abnormalities FUL L A N EUP LO IDY
A sex chromosome abnormality is not an uncommon discovery at prenatal diagnosis, with an overall incidence of 1 in 250–300 (Linden et al., 2002). The main conditions are XXY, XXX, XYY, and 45,X. As Boyd et al. (2011) write: “The importance of providing parents with accurate information about the frequency of the diagnosis and the variability of the condition on the basis of outcomes from unbiased population-based follow-up studies on the specific chromosome abnormality cannot be overemphasized.” Two of these aneuploidies (XXY and 45,X) may be firmly predicted in terms of an abnormality of development of the reproductive system. Children with Klinefelter and 45,X Turner syndrome will with near-certainty be infertile. For those couples deciding to continue a pregnancy, Robinson et al. (1986) offer a useful commentary. Parents of children predicted to be infertile might feel a sense of loss—a “sadness and regret about their child’s anticipated loss and about their own loss of grandchildren” and “concern about their children’s wholeness and, by extension, their own.” Parents may take some comfort from knowing that infertility is by no means an uncommon problem in the general population, and further comfort from the advice that recent advances in artificial reproductive technology may now enable the infertility to be overcome, in some individuals. The picture for intellectual and psychological functioning is less predictable. Earlier adult studies defining a strong association with mental deficiency and psychological disturbance were contaminated by ascertainment bias (and counselors’ personal experience may have been more with those children whose problems were sufficiently severe that they had come to medical attention). Children identified
in newborn populations screened for cytogenetic abnormalities and subsequently followed up constitute a group unbiased in their ascertainment, although perhaps subject to other but less important biases (Puck, 1981). Data from the study of such children in several American and European cities, followed from infancy through childhood, adolescence, and young adulthood, have now given a reasonably clear picture of the natural history of the more common sex chromosome aneuploidies (Linden et al., 2002). In general, the IQ averages 10–15 points below that of the siblings. Hook’s (1979) early proposition has held up: some sex chromosome aneuploidies influence brain function in such a way that the development of intellectual capacity, emotional maturity, and speech and language skills are affected to some extent; but none of these effects necessarily occurs, none is specific to sex chromosome aneuploidy, and some may be amenable to corrective intervention. There is considerable overlap with the XX and XY population. Ratcliffe (1999) and Bender et al. (2001) provide long-term follow up data, well into adulthood. Bender et al. followed eight 45,X, ten 47,XXX, and eleven 47,XXY individuals through to an age range of 26–36 years, using siblings as controls, and noted the IQs of the aneuploid groups to be considerably less compared with the sibs. Nevertheless, the variation is wide, and these authors emphasize the point that “sex chromosome aneuploidy does not exert its influence in a vacuum, but rather interacts with the host of other genetic and environmental influences that collectively guide human development.” Children with sex chromosome aneuploidies seem more susceptible to either the good or the bad effects of a stable or of a dysfunctional family setting than do their 46,XX and 46,XY siblings (Stewart et al., 1990; Bender et al., 1995). Children identified at prenatal diagnosis, a group biased toward higher socioeconomic status, may do better academically and socially than the cohorts followed from birth, although it was nevertheless true in the study of Linden and Bender (2002) that these children had “a strong risk for developmental problems, particularly for learning disabilities . . . [albeit that] these problems were not often severe.” There may, however, be an increased risk for psychosis in childhood and adulthood (Kumra et al., 1998). If a couple decides to continue the pregnancy, what should they say to others? Should the family know, should they tell friends, and should school personnel be aware? And when should the child Chromosome Abnormalities
•
477
learn about his or her chromosomal condition? Linden et al. (2002) have considered these questions, and in general make a case for openness within the family, but see no need, indeed potential disadvantage, for those outside to be told. We next outline the predicted outlook for the more commonly encountered sex chromosome aneuploidies. Attention is paid mostly to gonadal function and to intellectual and social development. X X Y ( K LIN EF ELTER SY N DROM E )
Almost certainly, the child becomes an infertile adult, although in recent times testicular sperm extraction with in vitro fertilization (IVF) has enabled a small number of men to become fathers (p. 224). Penile size is usually normal; the testes will be small. Androgen deficiency can be managed by replacement therapy with testosterone. It may be that treatment induces a more masculine body habitus, improved self-esteem, vitality, ability to concentrate, and sexual interest (Nielsen, 1990; Winter, 1990). Gynecomastia may be present, transiently, in some 50%; if it persists, it can be treated surgically. Verbal IQ is reduced by some 18 points, and performance IQ by 11 points (Leggett et al., 2010). Learning difficulty at school is to be expected. Of 13 XXY boys studied by Walzer et al. (1990), 11 had persistent reading and spelling problems. Bender et al. (1993) note that a deficit in verbal fluency and reading is “the most homogeneous and consistent cognitive impairment found in any sex chromosome abnormality group,” and this may reflect a specific dysfunction of the left cerebral hemisphere. Specific characteristics included a lowered level of motor activity, a pliant disposition, and a cautious approach to new situations; thus, in the classroom setting, they are perceived as “low-key children, well liked by their teachers, and presenting few behavioral management problems.” Leggett et al. (2010) conclude that these boys “do not usually have major problems with social interaction and adaptation, although they may be timid and unassertive.” Speculatively, the neural substrate of this passivity may reside in an underdevelopment of the amygdala, a brain nucleus that underpins aspects of social processing (Patwardhan et al., 2002).
Six Danish XXY boys were followed from birth to age 15–19 by Nielsen and Wohlert (1991), and all but one needed remedial teaching. Their career plans were carpenter, draughtsman, gardener, unskilled laborer, mechanic, and undecided. Stewart et al. (1990) comment that “XXY boys are unlikely to reach a level of personal and social development that is consistent with their family background.” Ratcliffe (1999) commented upon a rate of psychiatric referral being above that of male controls (26% cf. 9%), with the neurotic score (not the antisocial score) being higher. (She also notes anecdotal mention of men from a Klinefelter clinic with professions including physician, engineer, minister, and accountant.) In a summary of psychosocial adaptation from several studies, recurring adjectives to describe the XXY personality were shy, immature, restrained, reserved. In the Denver study, 11 young adults with XXY “appeared to have met the demands of early adulthood with fair success, although slightly less well than did their siblings”; they appeared to have a diminished insight into their own psychology (Bender et al., 1999). Their mean IQ of 91 compared with 109 in normal male sibling controls. We have noted earlier the ameliorative effect of growing up in a stable and supportive family.10 XXX
A full literature review of the XXX syndrome is provided by Otter et al. (2010), and the reader will find this helpful. Physical development of the XXX female is generally unremarkable, although there is a tendency toward tallness. Gross and fine motor skills are likely to be somewhat impaired, and children are awkward and poorly coordinated. Pubertal development and fertility appear, for the most part, uncompromised. In a very few, genitourinary malformations (ovarian, uterine, renal, bladder) are recorded, of which the karyotype may or may not have been causal (Haverty et al., 2004; Linden and Bender 2004). It is the neural substrate in which the important vulnerability applies (and which may reflect a reduced rate of cell cycles during neurogenesis; Otter et al., 2010). Thus, major concerns in childhood relate to intelligence and language development and poor self-confidence, and, in adulthood,
10 Besides detection prenatally, the condition may be screened for at different stages of postnatal life. Herlihy et al. (2010) use Klinefelter syndrome as an exemplar of how the pros and cons of diagnosis versus nondiagnosis may be assessed, at different times of life.
478
•
P R E N ATA L D I A G N O S I S
psychosocial maladjustment and, occasionally, frank psychiatric disease. Full scale and verbal IQ is reduced by some 10–20 points. Language comprehension and use of speech are impaired in over half the cases. Learning difficulty is likely and many will benefit from additional remedial teaching, but few require education outside the mainstream. In one small study of 11 girls, 9 needed special education intervention, and one was placed in a class for retarded children (Bender et al., 1993). Harmon et al. (1998) and Bender et al. (1999) reported a longer follow-up in these young women, into adolescence and young adulthood, and documented difficult adaptation to the stresses of life. On a measure of social adjustment (in work, leisure, family, marital, parental), the XXX women scored significantly less well than their sisters. Their mean IQ was 82 (cf. sisters, 103). However, Ratcliffe (1999) described most XXX young women in the Edinburgh survey as “physically attractive, and displaying a common sense attitude that counterbalanced their low educational achievements” (and relieved to be free of the pressure they had felt while at school). The observations in the similar study of Rovet et al. (1995) were more promising, although, as Harmon et al. point out, this was a group from a higher socioeconomic stratum, and presumably both genetic and environmental factors would have been more favorable. An XXX girl who might otherwise have had an IQ of 130 can yet do well in spite of a reduction to 110; to the contrary, a drop from 90 to 70 would be a considerable handicap. Many counselors will know from their own experience how variable can be the phenotype. XYY
The multicenter prospective study documented in Evans et al. (1990) reviewed progress in 39 boys and young men. The particular physical attribute of the XYY male is increased stature. Sexual activity is normal, and fertility is apparently uncompromised. Motor proficiency may be impaired. While the IQ is in the normal range, it is usually lower than those of sibs or controls, and about half of XYY boys have a mild learning difficulty, and may display poor attentiveness and impulsivity in the classroom. It may be that the aneuploidy causes a minor and subtle impairment of neurologic maturation, leading to some features of minimal brain dysfunction (Theilgaard, 1986). The vignettes from the series of Ratcliffe et al. (1990) of 10 Scottish subjects who
had left school give an idea of what XYY young men are capable of: one ran a market stall, two are chefs, and the others were a private in the army, a waiter, a supermarket assistant, a video shop assistant, a technician, a laborer, and one training as a painter and decorator. In a cohort of children aged 8–16 selected for the XYY karyotype having been diagnosed prenatally, and of higher socioeconomic status, a considerable range in academic ability was observed, with most coping satisfactorily, and IQs ranging from 100 to 147 (Linden and Bender, 2002). Perhaps the major concern is in psychosocial adaptation. These boys can have a low frustration tolerance, and some are prone to temper tantrums in childhood progressing to aggressive behavior in teenage and may need help to learn to cope with this. They may find it difficult to “read” social situations, and antisocial behavior is more common (Ratcliffe, 1999). The functioning of the family may be as much an ingredient as the karyotype in psychosocial development. Fryns et al. (1995) identified 50 XYY males among 98,725 patients referred for chromosomal analysis, and they note that this fraction of 50/98,725, approximately 1/2000, is very close to the newborn incidence, and thus drew a conclusion that the XYY phenotype differs little from the norm. They do, however, acknowledge a high (86%) risk for psychosocial pathology in those XYY males with concomitant borderline intelligence or frank mental deficiency. In Ratcliffe’s follow-up report into adulthood (1999), some disconcerting data are noted, not incongruent with the conclusions of Fryns et al. Psychiatric referrals were five-fold compared with male controls (47% cf. 9%), and the rate of criminal conviction four-fold, the mean IQ of those convicted being lower than those who were not (although most offences were minor, and against property rather than persons). 45, X (T UR N ER SYN D R O M E)
Unlike the foregoing aneuploidies, monosomy X has a very high in utero lethality, peaking at around 12 to 15 weeks gestation. Spontaneous abortion follows amniocentesis-detected 45,X in three-quarters of cases (Hook, 1983). But some survive pregnancy and are born as infants with Turner syndrome. Robinson et al. (1990) note that “variability among 45,X girls is considerable; and precise predictions about any child’s prognosis are not possible.” They also emphasized that “a supportive environment Chromosome Abnormalities
•
479
that provides stimulation and encouragement is of considerable importance.” These traits comprise the core phenotype (and a full description is given in Sybert, 2005): • Gonadal failure with infertility is almost certain (Lippe, 1991). In the survey of Sutton et al. (2005), infertility was seen, by the women with Turner syndrome themselves, as the most concerning component of the phenotype. Classically, a spontaneous onset of puberty, with breast development and onset of menses, has been regarded as being very infrequent, although Pasquino et al. (1997) proposed that the fraction who enter a spontaneous puberty may be as high as 9%, and they suggest that earlier figures may have been biased downward by a policy, previously, of not karyotyping short girls who had had an onset of menstruation. Childbearing via ovum donation may be successful in some cases (Chapter 13). Pavlidis et al. (1995) reviewed sexual functioning in women with Turner syndrome and suggest strategies to avoid possible difficulties. • Stature will be short. In a study of adult Danish women with Turner syndrome, never having had growth hormone therapy, the average height (with standard deviation) was 147 cm ± 7 cm (4 feet 10 inches ± 2½ inches) (Gravholt and Naeraa, 1997), which may be a little taller than in some other populations. A useful increment can be achieved with growth hormone treatment. • Neuropsychological functioning is impaired. The IQ is reduced compared to siblings. At longterm follow-up in the Denver cohort (Bender et al., 1999), nine young women with 45,X had a mean lower IQ (85) compared with normal female sibling controls (104). Their educational achievements were, however, better than those of the XXX women from the same study: eight were high school graduates, and five had college degrees. In one notable case, Reiss et al. (1993) report monozygous twins, one nonmosaic 45,X and the other 46,XX, the former’s performance IQ being 18 points less than her sister, but the verbal IQs practically the same. Psychological assessment indicates a particular vulnerability in social adaptation (Bender et al., 1999). Reiss et al. (1993) review aspects of the cognitivebehavioral phenotype and correlate the specific feature of difficulty with visual-spatial appreciation with a lesser volume of the right parietal cerebral cortex. Romans et al. (1998) confirmed and extended this appraisal in a study of 99 subjects with Turner syndrome, in whom they identified diminished abilities on measures of spatial and perceptual skills, 480
•
P R E N ATA L D I A G N O S I S
visual-motor integration, recognition of facial expressions associated with a particular affect, visual memory, attention, and executive function (the ability to plan, organize, monitor, and execute multistep problem-solving processes); the amygdala (see also XXY, earlier) may be the vulnerable neural substrate in this respect (Burnett et al., 2010). These traits are not improved by taking estrogen (Ross et al., 2002). • Certain physical defects are associated, of which the major are neck webbing and coarctation (narrowing) of the aorta. • Morbidity in adult life is increased (Gravholt, 2001; Swerdlow et al., 2001). Certain common diseases are more frequently seen: obesity, both insulin-dependant and insulin-resistant diabetes, hypothyroidism, heart disease, hypertension, stroke, and liver cirrhosis. Weakness of the bones (osteoporosis) implies a risk for fracture. There may be a place for ongoing hormone replacement therapy. There is a possibility that Y-chromosome material may be present even if the karyotype is apparently nonmosaic 45,X. Huang et al. (2002) reviewed 74 cases of 45,X diagnosed prenatally, most having been ascertained via, or discovered with, abnormal fetal ultrasonography. Of six with normal ultrasonography, three showed a male genital phenotype. The explanations, upon more detailed analysis, were as follows: in one, a segment of Yp was translocated to a chromosome 14, shown on FISH with an SRY probe, and in the other two, there was low-level mosaicism for an idic(Y) marker. Normal male children were born. Some women with Turner syndrome who are 45,X on karyotyping may actually show Y sequences on molecular study, and these women do have a greater risk for gonadoblastoma (Mendes et al., 1999). SE X CH R O M OS O M E PO LYS O MY
This category is to be seen as quite separate from 47,XXX and 47,XXY. Linden et al. (1995) review the phenotypes of 48,XXXX, 48,XXXY, 48,XXYY, 48,XYYY, 49,XXXXX, 49,XXXXY, 49,XXXYY, 49,XXYYY, and 49,XYYYY, and 48,XXYY, 48,XXXY, and 49,XXXXY are also outlined in Visootsak and Graham (2006). In each, variable intellectual compromise is characteristic. While the authors’ comment is well taken that the current perception of the seriousness of phenotypic abnormality may have been overstated due to ascertainment bias, and indeed they describe normal (but low) IQs in some of the 2n = 48 karyotypes, it remains true that most
have substantial handicap due to intellectual deficit and abnormal behavior (Cammarata et al., 1999). The very rare 49,XYYYY karyotype is described in Paoloni-Giacobino and Lespinasse (2007); almost as rare is 49,XXXXY, concerning which Peitsidis et al. (2009) review prenatal diagnosis, noting that nuchal thickening is a frequent observation. IQ in both these pentasomies is very low. XX Male. Discordance of 46,XX karyotype and male genital phenotype, as seen on ultrasonography, allows prenatal diagnosis of the XX male (TrujilloTiebas et al., 2006). Other possible diagnoses need consideration, including congenital adrenal hyperplasia with virilization of a female fetus (Lonardo et al., 2009); of course the possibility of maternal cell contamination will need to be excluded. Normal intelligence and stature are predicted, but there will be testicular deficiency with infertility (Margarit et al., 1998). X AND Y C H RO MOS O M E M OSA I CI SM
True mosaicism involving the sex chromosomes seen at prenatal diagnosis presents a challenge in interpretation, and skilled ultrasonography, with respect to external genital anatomy, is central in determining the fetal gender. The problem is that the tissue analyzed at prenatal diagnosis may or may not reflect the distribution in the gonad. The presence of a Y chromosome in at least some gonadal tissue—or to be precise, the presence of the Y-borne SRY gene—will promote testicular development, which might or might not be complete, and which might or might not secrete male-inducing hormones. Thus, we may observe gender states from normal (although possibly infertile) female, through Turner-like female, genital ambiguity, mixed gonadal dysgenesis, even ovotesticular disorder of sex development (p. 336), to male with incomplete pubertal development, and to normal (although often infertile) male. XX/XY. This is usually pseudomosaicism, resulting from the growth of maternal cells in a 46,XY pregnancy (Worton and Stern, 1984). (Obviously, such pseudomosaicism would normally be undetected if the fetus is female.) Level III XX/ XY mosaicism, curiously enough, is most likely to indicate a phenotypically normal female fetus in which the XY source is unknown, particularly when the XX cells predominate. A male “vanished twin” is a theoretical possibility (Worton and Stern, 1984),
and indeed a quite plausible explanation, given the frequency with which a twin pregnancy prior to 7 weeks is followed, some months later, by the birth of a singleton baby (Sampson and de Crespigny, 1992). Analysis of placental membranes after delivery in a pregnancy from which one twin has “vanished” can often reveal a fetus papyraceous or a remnant empty sac (Nerlich et al., 1992). One can imagine tissue of the (male) twin remnant having been, by chance, in the path of the amniocentesis needle that sampled cells from the remaining (female) fetus. A girl born following such a prenatal diagnosis (Hunter et al., 1982) was followed through to mid-adolescence, and her development was entirely normal (A. G. W. Hunter, personal communication, 2002). A similar case was studied by I. Hayes and A. George (personal communication, 2009), with an XX:XY ratio of 90:10 on FISH of uncultured amniotic fluid, although nonmosaic 46,XX on cultured cells; ultrasonography indicated female external genital morphology. Following the birth of a normal girl, examination of seven sites from the placenta, and one site each from the cord and sac, all revealed a nonmosaic female sex complement, as did the cord blood sample. A true fetal XX/XY karyotype is rare indeed, and it is more likely due to the fusion of two gametes; that is, chimerism (but other mechanisms exist; see Chapter 20, section on “Ovotesticular Disorder of Sex Development”). Presumably depending upon the gonadal distribution of XX and XY cells, the genital anatomy will be male, female, or in between. Malan et al. (2007) report XX/XY chimerism at prenatal diagnosis, the child (subjected to pelvic ultrasonography) proving to be an apparently normal girl. Ovotesticular DSD, with imperfect or ambiguous genital anatomy, has been recorded from an XX/XY amniocentesis result, with the same karyotype demonstrated in the child (Amor et al., 1999; Chen et al., 2005c, 2006f; Malan et al., 2007). Yaron et al., (1999) had a case presenting at amniocentesis, with normal male morphology on ultrasound. The XX/XY mosaicism was confirmed on a second amniocentesis, and, in due course, on the normal male newborn infant (including on genital skin). Amor et al. note the point that intellectual compromise is not to be anticipated. Hughes et al. (2006) provide guidelines on management for children with intersex conditions. Infertility is predicted; but remarkably enough, one XX/XY man has fathered a child, following IVF with retrieved sperm (Sugawara et al., 2005). Chromosome Abnormalities
•
481
X/XY. Patients coming to medical attention and found to have 45,X/46,XY mosaicism range in phenotype from females with classical Turner syndrome, through infants with ambiguous genitalia, to normal but infertile males (Telvi et al., 1999; Tho et al., 2007). A risk for gonadal tumor applies (Müller and Skakkebæk, 1990; Müller et al., 1999). By contrast, a normal male infant is the outcome in the considerable majority (90%–95%) of X/XY gestations detected at prenatal diagnosis—in other words, cases whose ascertainment was unbiased—and going through to birth (Hsu, 1994; Huang et al., 2002). Fertility, however, is likely to be compromised. Van den Berg et al. (2000) report a case in which nonmosaic 45,X was diagnosed at short-term CVS, with a nonmosaic 46,XY karyotype seen on long-term culture. Subsequent amniocentesis revealed a true 45,X/ 46,XY mosaicism. Post termination, fetal testing showed X/XY mosaicism in all tissues sampled (including gonads). Of 14 pathology studies on fetuses post termination in Chang et al. (1990), two were found to have ovotestes and one had a “precancerous” lesion. Other Sex Chromosome Mosaicism. The karyotypes most frequently seen are 45,X/ 46,XX, 47,XXY/46,XY, 45,X/47,XXX, and 47,XXX/ 46,XX. It appears that the considerable majority of cases of true sex chromosomal mosaicism of these types are associated with concordant (Y→male, no Y→female) and normal genital development (Hsu and Perlis, 1984; Wheeler et al., 1988). In X/XX, X/XXX, XXX/X/XX, and XXX/XX mosaicism IQ is not discernibly affected; verbal IQ may be slightly lowered in XXY/XY (Netley, 1986; Bender et al., 1993). Huang et al. (2002) reported a case each of X/XXX and XXX/X/XX mosaicism diagnosed at amniocentesis, the former pregnancy producing a newborn with features of Turner syndrome, and the other a normal female. X/XX. Koeberl et al. (1995) record 12 cases of 45,X/46,XX mosaicism detected at amniocentesis, with the percentage of 45,X cells in 10 of these being in the region of 20%–70%. Postnatal studies (blood and/or skin) confirmed the mosaicism in nine (at a lesser percentage in all but one), while in three of the children no 45,X cells were seen. None showed growth retardation postnatally and in none would a clinical suspicion of Turner syndrome have arisen. Two cases of presumed early ovarian dysfunction, one of these also having urogenital anomalies, 482
•
P R E N ATA L D I A G N O S I S
might reflect an effect of the karyotype; it is possible some of the remaining cases could also manifest abnormal ovarian function at a later age. The abnormal neurology in one of the twelve is of uncertain significance. Koeberl et al. comment that “the prenatal diagnosis of 45,X/46,XX is not necessarily benign.” Hsu (1996) and Sybert et al. (1996) debate the validity of previous larger series of X/XX prenatal diagnoses. A clear point forthcoming is that data from longer term follow-up are desirable, given that a functional gonadal component of the syndrome might not manifest until well into adolescence or adult life. In 2002, Huang et al. reported their experience with 17 cases of X/XX mosaicism at amniocentesis. The ratios of X to XX cells ranged from 2:23 to 12:3. One case with intrauterine growth retardation (ratio 6:12) terminated in stillbirth, while the remaining 16 had normal ultrasonography. Of the eight cases continuing to term and for which information was available, two liveborn babies had the features of Turner syndrome (ratios 7:10 and 3:14), with the mosaicism confirmed postnatally in one of these. The remaining six (ratios ranging from 3:15 to 12:8) “reportedly had a normal female phenotype.” To quote Huang et al. “the percentage of 45,X cells in amniocytes does not seem to be an indicator of pregnancy outcome as there was considerable overlap between cases with normal and abnormal outcome.” In a unique case of a monozygous twin pregnancy, one fetus showed nuchal swelling and the other appeared normal (Gilbert et al., 2002). Fetal blood sampling showed low-grade 45,X[2]/ 46,XX[23] mosaicism in the former, and a normal 46,XX karyotype in the latter, in contrast to postnatal skin fibroblast karyotyping results of nonmosaic 45,X and 45,X[2]/46,XX[78], respectively. X/XXX/XX and X/XXX. One reported case of X/XXX/XX mosaicism illustrates the difficulty in extrapolating the distribution of cell types from one tissue to another (Schwartz and Raffel, 1992). Amniocentesis gave the proportions 16:20:64, respectively. Cord blood gave similar findings, although in placental tissue (chorion) the percentages were 2:41:57. The baby appeared normal. Sybert (2002) reviewed hers and others’ data, and concluded that about 60% of girls with X/XXX/XX and X/XXX could be predicted to have short stature, and that “it is fair to suggest that residual ovarian function is possible and to caution that premature ovarian failure is common.”
X/XYY and X/XYY/XY. The X/XYY and X/ XYY/XY mosaic states are (necessarily) abnormal in postnatally ascertained cases, but prenatally diagnosed cases have consistently manifested an apparently normal male genital phenotype, albeit that the mosaicism may be confirmed in the child subsequently born (Pettenati et al., 1991; Hsu, 1994). Presumably according to the distribution of X and XYY tissues, the gender in X/XYY mosaicism can be of either sex, or there can be ambiguity, these three states documented in one of the earliest reviews (Mulcahy et al., 1977). There is a tumor risk, and gonadoblastoma was identified at gonadectomy in a virilized female with mixed gonadal dysgenesis (Gibbons et al., 1999). Infertility is likely, but may be treatable (Dale et al., 2002). It is hypothetical whether the XYY line, if involving the brain, would determine an intellect and psyche of the XYY “syndrome” (earlier). STRUC TUR ALLY A B NORM A L SE X C H RO MOS O ME
X-Y Translocation. The most common form of the t(X;Y) has the X breakpoint at or distal to Xp22, and the Y breakpoint at Yq11.2. The intact sex chromosome may be an X or a Y chromosome, and the two states differ as follows. 46,X,der(X)t(X;Y): A de novo X-Y translocation would be expected to herald a female child, who will likely be short, 150 cm or less in height as an adult ( Joseph et al., 1996; Speevak et al., 2001). The site of the breakpoint can be pinpointed with probes for two loci (steroid sulfatase, Kallmann syndrome) in Xp22.3; if these loci are present on the der(X)t(X;Y), intelligence and fertility may be intact, and other defects are unlikely. A few de novo cases have been associated with major defects, presumably due to a marginally more proximal Xp breakpoint, with the deletion of crucial genes. 46,Y,der(X)t(X;Y): If the intact sex chromosome is the Y, the child is expected to be male. If the probes noted earlier are present, the phenotype is likely to be confined to short stature and infertility. A more extensive loss of loci might determine a nullisomy that would cause important malformation and be lethal in utero. (Unbalanced X-autosome and Y-autosome translocations are dealt with in the section earlier on autosomes.) Other rare types include dicentric X;Y translocations, and der(X) and der(Y) chromosomes with a range of p and q arm breakpoints on X and Y
(Hsu, 1994). The phenotypes are male if SRY is present, and otherwise female. Infertility is typical, and, in the male, short stature. In the der(Y) case, in which there may be an effect of functional X disomy, genital anomaly and other malformation is common, as is mental defect. A detailed case is described in Ghosh et al. (2008), in which the recognition of an ultrasound brain anomaly at 21 weeks led to amniocentesis with the discovery of a de novo 46,X,der(Y) t(X;Y)(p22.13;q11.23). The Yqh region was replaced by Xp material, which thus existed in the functionally disomic state. X Chromosome Deletion. The possibility of an inherited X-autosome translocation should be checked by doing the mother’s karyotype. Alternatively, it may transpire that the mother has the same karyotype. X chromosome deletions in the female, 46,X,del(Xp) or 46,X,del(Xq), predict the possibility, but not the certainty, of an incomplete form of Turner syndrome and/or premature ovarian failure (Fitzgerald et al., 1984; Veneman et al., 1991). Brown et al. (2001) describe a mother, of tall stature (5 feet 10 inches), having a prenatal diagnosis of del(X)(q22q26); she herself had the same karyotype, and “the parents took comfort in the observation that in the mother the deletion had no apparent phenotypic effect.” A normal baby girl was born. Mother and daughter showed completely skewed X-inactivation, the abnormal X being consistently inactive. In the male, the 46,Y,del(X) state would be nonviable for all but the very smallest deletions, and major abnormality would be probable for those pregnancies that might be viable. X Chromosome Duplication. De novo X chromosome duplications, 46,X,dup(X), in the female might have been thought to be of minimal effect (a partial XXX syndrome) due to selective inactivation of the abnormal X. This is sometimes but not necessarily the case, and abnormal phenotypes, often including genital defect, are not infrequently observed. Zhang et al. (1997) provide detail according to the extent and site of the duplication in a review of postnatally diagnosed cases. Tihy et al. (1999) describe an infant girl with a de novo dup(X) (q22.1q25) who had physical and neurodevelopmental defects, in spite of the X-inactivation pattern (at least on peripheral blood) showing consistent inactivation of the dup(X). Functional disomy, at least for part of the segment, may contribute to the Chromosome Abnormalities
•
483
abnormal phenotype in such cases (Armstrong et al., 2003; Tachdjian et al., 2004). Normality has been reported with respect to an isodicentric X, idic(X)(q27), comprising practically a double copy of the X, identified prenatally, the abnormal chromosome being late replicating, and indeed one such child was “academically advanced and enrolled in a gifted and talented program”; in contrast, some postnatally diagnosed patients have presented a Turner-like clinical picture (Tsai et al., 2006). In the male, functional disomy for the duplicated segment would likely cause severe defects, often lethal in utero. The tiny marker X seen in the 45,X/46,X,+min(X) karyotype may be considered in the same category as the tiny ring X syndrome, discussed later. Other Abnormal X Chromosomes X chromosome abnormalities are characteristically seen in the mosaic state, the other cell line typically being 45,X. Mosaicism with a large ring X or an isochromosome for the X long arm, 45,X/46,r(X) and 45,X/46,X,i(Xq), respectively, would lead to variant Turner syndrome. An isochromosome for the X short arm, i(Xp), would probably always be lethal, since there would be a functional Xp trisomy (Lebo et al., 1999). As with the small ring X syndrome (see following), a marker X that lacks XIST is associated with phenotypic abnormality (Tümer et al., 1998). The “tiny ring X syndrome” with the karyotype 45,X/46,X,r(X) may have a functional X disomy and is typically, but not universally, seen with a severe phenotype of physical and mental defect, in some resembling Kabuki syndrome. The severity of the phenotype has been attributed to a functional X disomy, due to the ring lacking the XIST locus and thus not undergoing inactivation, although in rare instances this scenario may not apply (Migeon et al., 2000; Rodríguez et al., 2008). Similarly, in the male with the tiny ring as a supernumerary chromosome, usually as 46,XY/47,XY,+r(X) mosaicism, the clinical picture is typically abnormal, and in some severely so (Baker et al., 2010). Chen et al., (2006e) report an exception, from the prenatal diagnosis at amniocentesis of 46,XY[17]/47,XY,+mar[6], the
marker turning out to be a very small XIST-negative r(X). The infant boy, on whose blood the proportions of the two cell lines were similar to the amniocentesis findings, was normal physically and developmentally, on follow-up to 1 year of age. These phenotypic differences may reflect the fraction and distribution of the abnormal chromosome. A BNO R M A L Y CH R O M OS O M E
Hsu (1994) lists several possibilities: Yq– of various extents (excluding normal Yq variation), Yp–, r(Y), and isochromosomes or isodicentric chromosomes, written variously as i(Yp), idic(Yp), i(Yq), and idic(Yq). Concomitant 45,X mosaicism is very often observed, and this complicates prediction; ultrasonographic assessment of external genital morphology may be useful. Intactness of Yp with loss of Yq loci, and in particular the AZF spermatogenesis loci (Fig. 6–1), is associated with male infertility. Absence of SRY leads to female development, and loss of other Yp loci determines a Turner syndrome phenotype. In 45,X/46,X,der(Y) with Turner syndrome there is a risk for gonadoblastoma (Atkins et al., 2000). The outcomes in one prenatal diagnostic series are set out in Table 27–3. Y Isochromosome. The least rare of these rare conditions is the Y isochromosome, or isodicentric chromosome, usually seen as 46,X,i(Y)(p10) or 46,X,i(Y)(q11), in which the essential imbalance is a double dose of Yp material, and absence (or nearly so) of Yq.11 As noted earlier, the condition may be seen in both nonmosaic and (more usually) mosaic form, the latter with a 45,X cell line. The phenotype in postnatally identified cases has ranged from sterile but otherwise normal male, through female somewhat resembling “testicular feminization,” to actual genital ambiguity (Bruyère et al., 2006; DesGroseilliers et al., 2006). In contrast, the outlook from unbiased (that is, not following an abnormal ultrasound) prenatal diagnosis is markedly in favor of normal male physical development, albeit that infertility will be very probable, and indeed, practically certain (p. 391). Ultrasonography is crucial: if this indicates male genitalia, a normal male
11 An interesting question, not entirely theoretical in the present context, is what extrapolation, if any, can be made from the XYY syndrome, in which there is a double dose of Yp, but of course also of Yq material. Neas et al. (2005) suggest that trisomy for the pseudoautosomal region PAR1 might lie behind aspects of the cognitive phenotype in the XXX and in some i(Y) karyotypes; and the same might apply to XYY.
484
•
P R E N ATA L D I A G N O S I S
phenotype is to be anticipated. Willis et al. (2006) reviewed 15 cases, with follow-up from 4 months to 9 years: all but one had presented as normal males, and “development has been normal in all cases where follow-up was reported.” A similarly optimistic interpretation comes from Bruyère et al. (2006): in a series of 12 cases from these authors, all 9 in which diagnosis had been unbiased, and the pregnancies continued, led to births of normal males, and normal development in those who were further followed up. While a question about cognitive development is not entirely settled11 (Tuck-Muller et al., 1995; Neas et al., 2005), and few reports give follow-up into adolescence or adulthood, at least anecdotally, some do well. Y Ring. Layman et al. (2009) report their own cases and review the 45,X/46,X,r(Y) karyotype, as identified in males in whom testes were descended. Variable short stature and gonadal failure were typical. These authors note the confounding factors, in terms of predicting phenotype at prenatal diagnosis,
of the bias toward genital abnormality in postnatally identified infants, versus the frequent lack of follow-up in apparently normal males following a prenatal diagnosis, leading to a bias in the other direction. Three very rare Y scenarios may be mentioned. (1) The pericentric inversion Y is typically regarded as being a nonpathogenic variant (p. 173), although Gimelli et al. (2006) have reported a young woman with such an inversion, who presented with gonadal dysgenesis and bilateral gonadoblastoma. (2) A single case is recorded of a retarded man who had two copies of an idic(Y)(q12), conveying a tetrasomy of Yp; he rather resembled the picture of the XXXXY syndrome (Maas et al., 2005). (3) Mosaicism for a Yqh of different lengths, the shorter presumably having been derived from postzygotic deletion of the longer, has been identified prenatally in a very few cases (Cotter and Norton, 2005); a phenotypic consequence would seem unlikely, unless a Yq gonadal locus had been affected, which might theoretically compromise fertility.
Chromosome Abnormalities
•
485
This page intentionally left blank
PART EIGHT NOXIOUS AGENTS
This page intentionally left blank
28 gonadal cytogenetic damage from exposure to extrinsic agents
IN THIS CHAPTER we review what is known about the effects of some anticancer treatments, and of certain other therapeutic and environmental agents, that could conceivably have an injurious effect upon chromosomal distribution at gametogenesis, or which might cause chromosomal breakage or rearrangement in the cells of the gonad. In other words, the focus is on factors that might disturb the course of meiosis, or that might have clastogenic effects upon the chromosomes of gametocytes. We do not consider other categories of genetic damage. Given the inherent vulnerability of gametogenesis, a logical starting position might have been that any potential damaging agent should be presumed guilty until proven innocent. As discussed in Chapter 23, large fractions of sperm and eggs, in the vicinities of 10%–20%, are chromosomally abnormal, due to aneuploidies or structural change acquired, for the most part, during meiosis. If this is what happens naturally, if gametogenesis is so
susceptible normally, then surely would not agents known to compromise the integrity of the DNA and of the spindle apparatus (not to mention various artificial dietary and environmental exposures) compound the effect dramatically? Perhaps surprisingly, this seems not to be the case. Gametogenesis— provided the damage is not irreversible—often proceeds normally, or at any rate recovers, even in the setting of some heavy exposures, and no discernible increase in chromosomal abnormality is recorded in the subsequently born children. Nevertheless, if only on the pure grounds of what seems biologically reasonable and plausible, the question is not to be regarded as being closed. The fact that sperm chromosomes may, with certain agents, show an increased rate of cytogenetic abnormality is a more practical reason for maintaining a cautious view. We outline the observations and conclusions relating to a number of medical conditions, cancer and otherwise, and touch on some environmental and lifestyle factors. The listing is not exhaustive.
•
489
BIOLOGY AND EPIDEMIOLOGY Cancer Treatment A majority of children and young adults who receive modern cancer treatment survive. Some treatments cause sterility, but in quite a number fertility is unscathed, or at any rate subsequently recovers (Green et al., 2009, 2010). For those who are potentially capable of having children, the question arises: could there be an increased risk to have a child with a chromosomal abnormality? For most, in fact, the short answer may be, apparently not. Longer answers follow. The chemotherapeutic agents and radiation used to rid the body of cancer are essentially cellular toxins, some of which specifically target DNA or the mitotic apparatus. Thus, the starting hypothesis is that the chromosomes in exposed bystander tissues, and thinking in particular of the gonad, could be vulnerable. The fact that these treatments can damage chromosomes is well known, and this is actually the basis of one of the in vitro laboratory tests for ataxia-telangiectasia.1 Rapidly dividing cells are the most vulnerable to anticancer treatments (this being, of course, the rationale for their use). This would suggest, in theory, a susceptibility for spermatogenesis in the postpubertal male (millions of cell divisions daily), and a relative resistance in the prepubertal child (male meiosis yet to commence) and in oögenesis from infancy through menopause in the female (cell division in suspension). The direct assessment of gametic chromosomes offers insight. Sperm chromosome analysis can be done in men who have survived cancer treatment. Table 28–1 carries a review of 12 such studies and shows that several therapeutic regimens can cause sperm karyotypic defects.2 In practice, it is to the experience of the “therapeutic experiments” of oncological medicine that we mostly appeal: the in vivo observations of those who have survived their cancer, recovered from their treatment, and who have gone on to have, or to attempt to have, children. Have the children shown any excess of cytogenetic abnormality? The two largest studies are those from the Five Center3
collaboration in the United States and a nationwide survey from Denmark (Byrne et al., 1998; Winther et al., 2004). In the American study, 1062 survivors born in the period 1945–1974 were interviewed in 1980–1983, these persons having had several types of childhood cancer (Hodgkin’s disease, soft tissue sarcoma, and thyroid and central nervous system tumors accounting for the majority). In Denmark, a similar cohort of 4676 cancer survivors were compared with their 6441 siblings. The outcomes in the children were assessed. The rates of chromosome abnormalities in the offspring in these two studies did not differ significantly from those of their cancer-free siblings (Table 28–2).
SPECIFIC THERAPIES
Chemotherapy. Levy and Stillman (1991) and Arnon et al. (2001) review in detail the effects of various chemotherapeutic regimens upon fertility, in some of which data are also available concerning mutagenicity. The six classes of chemotherapeutic agents are the following: alkylating agents, cisplatin and its analogs, vinca alkaloids, antimetabolites, topoisomerase inhibitors, and “newer agents.” In broad terms, the relationship between type of drug and risk for gonadal damage is outlined in Table 28–3. Alkylating agents (cyclophosphamide and chlorambucil being major representatives), which have their damaging effect by adding an alkyl group to DNA, can, in the male, cause testicular hypotrophy, with oligospermia or azoöspermia. In follow-up into adulthood, the reproductive potential for males having been treated with an alkylating agent in childhood is considerably reduced, with a relative fertility of 0.4 (Byrne et al., 1987). Levy and Stillman review a number of papers, which offer a generally optimistic picture for girls in terms of pubertal development, but, as they point out, longer-term studies relating to the specific question of fertility are not so numerous. In one follow-up study, women actually had a relative fertility of 1.0 (Byrne et al., 1987). In adult women who have had chemotherapy with cyclophosphamide for Hodgkin’s disease or breast cancer, oöcyte depletion and ovarian failure
1 Radiation and bleomycin, both having potent DNA-breaking properties, cause lymphocyte chromosome rearrangements in normal, and considerably more so in ataxia-telangiectasia cells. 2 Male cancer patients may show abnormal sperm genetic studies ahead of having received any treatment, suggesting that there is a harmful effect of the malignant disease per se (Tempest et al., 2008). 3 Universities of Iowa, Kansas, and Texas, California Department of Health Services, Connecticut Tumor Registry.
490
•
NOXIOUS AGENTS
Table 28–1. Findings from 12 Sperm Chromosome Studies That Have Been Done in Respect of the Treatment of Certain Cancers, Showing Those Regimens That Are Associated, and Those That Are Not, with Sperm Chromosomal Abnormalities INCREASED FREQUENCY CHROMOSOME ABNORMALITIES CANCER
THERAPY
Cancer
PEB and D-act and CY-CH-V-MT Testic. ca. (non-sem.) PVB, PVB and PEB Rhabdomyosarcoma CYVADIC Ewing sarcoma VAC Wilms tumor RT ± D-Act Seminoma PVB Hodgkin’s MOPP, MOPP and RT Lymphoma MACOP-B Lymphoma MACOP-B Embryonal cancer PEB Embryonal cancer PEB Hodgkin’s NOVP Hodgkin’s Vinb. and RT Testicular cancer (non-sem.) PEB Testicular cancer (non-sem.) PEB
CYTOGENETICS
STRUCTURAL
NUMERICAL
Humster
+
+
Humster Humster Humster Humster Humster Humster Humster FISH FISH Humster FISH FISH FISH FISH
+ + + + – + – + – – + + + +
+ – – – – + – – + – +/– ++ ++ +
“humster” = pseudofertilization human-hamster test (p. 379). Testic. ca. (non-sem.) = testicular cancer, nonseminoma. Treatment regimens: CY-CH-V-MT, cyclophosphamide, chlorambucil, vinblastine, methotrexate; CYVADIC, cyclophosphamide, adriamycin, vincristine, dicarbazine; D-Act, D-actinomycin; MACOP-B, methotrexate, doxorubicin, cyclophosphamide, vincristine, prednisone; MOPP, nitrogen mustard, vincristine, procarbazine, prednisone; NOVP, novanthrone (mitoxantrone), vincristine, vinblastine, prednisone; PEB, cisplatin, etoposide, bleomycin; PVB, cisplatin, vinblastine, bleomycin; RT, radiotherapy; VAC, vincristine, adriamycin, cyclophosphamide; vinb., vinblastine. Source: From the review of De Mas et al. (2001).
Table 28–2. Outcomes from Two Large Studies of the Offspring of Childhood Cancer Survivors UNITED STATES
Numbers Offspring Chromosome abnormalities Down syndrome* Turner syndrome*
DENMARK
SURVIVORS
THEIR SIBLINGS
SURVIVORS
THEIR SIBLINGS
1062 2198 0.2% 0.1%
2032 4544 0.1% 0.1%
4676 2630 0.21% 1.07 1.32
6441 5504 0.21% 1.0 1.0
*U.S. figures = birth prevalence; Danish figures = relative risk. The gender of the affected parent of the DS and TS offspring in the Danish material suggested the nondisjunction event would more likely have been in the unaffected partner, and thus the relative risks listed above (in any event statistically nonsignificant) may misrepresent a cancer-related effect. The American data are reevaluated in Meistrich and Byrne (2002). Sources: From Byrne et al. (1998); Winther et al. (2004); and see text.
Table 28–3. Three Categories of Risk for Germ Cell Depletion, Based on the Drugs Used
The frequencies correlated with the estimated “bystander” testicular radiation (that is, the extent to which exposure extended beyond the target tissue).
DEGREE OF RISK
DRUG
Definite
Mechlorethamine, chlorambucil, procarbazine, busulfan, cyclophosphamide Doxorubicin, vinblastine, cytosine arabinoside Methotrexate, 5-fluorouracil, 6-mercaptopurine
Radioisotopes. Radioiodine is used in thyroid cancer, and in a review of 408 offspring of survivors in Ehrenheim et al. (1997), no increase in congenital malformations was noted, although in one case a 7/14 translocation, not further described (and not identified as de novo or familial), was detected at prenatal diagnosis. One case does not make a case, but it can be noted.
Probable Unlikely
Source: From Levy and Stillman (1991).
are documented (Familiari et al., 1993; Goodwin et al., 1999). Antimetabolites, alkaloids, and antibiotics (including methotrexate, vincristine, actinomycin D) seem not to cause compromise ovarian function when given alone or as combination therapy, but in conjunction with radiotherapy some will cause ovarian failure. Topoisomerase inhibitors affect the integrity of the mechanical apparatus of the meiotic chromosome, including the centromere and the microtubules of the spindle, and they also act directly upon the DNA; mouse studies with etoposide show an actual increase in sperm and zygote aneuploidies (De Mas et al., 2001; Marchetti et al., 2001). Multiagent chemotherapy is, as would be expected, more damaging, and as illustrated in a group of girls who had succumbed to their cancer, from the observations of ovarian histology post mortem (Nicosia et al., 1985). Radiotherapy. Fertility is diminished in females who have had radiation therapy to the abdomen, and there is an increased risk of obstetric complication. But their children appear to have no increased incidence of birth defects (Nicholson and Byrne, 1993). Martin et al. (1986b) studied 13 male cancer patients (mostly seminoma) at intervals up to 36 months after radiotherapy, in whom the doses of testicular radiation were estimated to be in the range 0.4–5.0 Gray. While most were azoöspermic in the first year following treatment, in those in whom spermatogenesis recovered, variable increases in sperm chromosome abnormalities were seen, averaging two-fold overall compared with controls, but with wide ranges. 492
•
NOXIOUS AGENTS
SPECIFIC DISEASES
Hodgkin’s Lymphoma. Treatment for Hodgkin’s lymphoma typically involves radiation to the chest and abdomen, and multidrug chemotherapy (e. g., MOPP, NOVP, ABVD; see abbreviations in footnote of Table 28–1). Spermatogenesis is compromised, and may or may not recover, although ovarian function typically is resistant or, if affected, more readily returns to normal (Marmor and Duyck, 1995; Papadakis et al., 1999). Some males have increased sperm aneuploidy rates before starting treatment, pointing to an effect upon spermatogenesis of the disease state itself. Tempest et al. (2008) review previous work on sperm chromosome analyses pre and post treatment, and report their own findings. In their material, chromosomes 13, 12, X, and Y were analyzed by fluorescence in situ hybridization (FISH), in very large numbers of sperm, from five patients age 19–36 years, and sampled at 0, 6, 12, and 18–24 months. Disomies and nullisomies for these chromosomes occurred at a higher frequency than in controls, albeit that the absolute amounts were small, at fractions of a percent. The aneuploidy rate declined to pretreatment levels by 18–24 months, although the authors retain a reservation that a 24-month cutoff could not necessarily be taken as a timeframe beyond which no increased risk would exist. The foregoing work on sperm notwithstanding, actual observed reproductive outcomes from previously treated patients are the proof positive, and the observations are substantially reassuring. Aisner et al. (1993) interviewed 35 women and 25 men who had had 68 living children. There was no increase in spontaneous abortions or congenital abnormalities. Six women in the study of Papadakis et al. (1999) had had eight normal children. Similar findings were obtained by Swerdlow et al. (1996), from
11 men and 16 women who had had a total of 49 children. Chromosome analyses were done in 45 of these children, and all were normal, except for one child with trisomy 21, the additional chromosome having been transmitted from the other parent. These authors concluded that “offspring of patients treated in adulthood for Hodgkin’s disease are not at greatly raised risk of genotoxic or other adverse outcomes as a consequence of their parent’s treatment.” This provides a counterpoint to the concern raised by the in vitro sperm studies discussed earlier. It would be discretionary whether prenatal diagnosis might be chosen in a pregnancy conceived either by using pretreatment stored sperm or in a natural pregnancy following recovery of spermatogenesis. Childhood Acute Lymphoblastic Leukemia. About half of males treated for acute lymphoblastic leukemia (ALL) will suffer sterility, following the typical regimen of alkylating agents. López Andreu et al. (2000) undertook sperm analyses in a group of 22 childhood leukemia survivors, they at the time being teenagers or young adults. Five were azoöspermic or severely oligoasthenozoöspermic. The ovary, on the other hand, is more resistant to a permanent effect of alkylating agents. In those in whom fertility recovers, there has been no indication of any increase in the rates of congenital malformation in the offspring (Levy and Stillman, 1991). Kenney et al. (1996), in a study based on 140 children born to ALL survivors, record one case of trisomy 13, but overall there was no significant difference in malformation rates between the children of survivors (3.6%) and those of their sibling controls (3.5%). Wilms Tumor. In a review of Wilms tumor survivors, Byrne et al. (1998) documented a particular risk for the female to have children with adverse outcomes, but this effect is apparently due to a damaging influence of radiation on the uterus with a secondary deforming effect on the fetus, or possibly reflecting a transmitted genetic defect that had been responsible for the mother’s original tumor as a child. No malformations were recorded in the offspring of 19 children of male survivors, this being, admittedly, a small number. Testicular Cancer. This is typically a cancer of younger men. The treatment is usually surgical removal of the affected testis, and chemotherapy with such agents as cisplatin, etoposide, and bleomycin. Fertility is maintained in some, although
persisting oligospermia is a frequent observation, especially in those having had high-dose cisplatin (Stephenson et al., 1995). Sperm aneuploidy rates are increased, using certain chromosomes as surrogates, but the normality rate per autosome usually remaining above 99½%, and per sex chromosome, above 99%, in each case barely different from control data (Tempest et al., 2008). A risk to produce aneuploid sperm may diminish with the passage of time after the chemotherapy. Martin and colleagues (1997, 1998, 1999) studied four men variously before, during, and some 2–13 years after treatment with the three drugs noted. These men, after successful treatment, had six more children, all normal. Another eight normal children are recorded in the series of Stephenson et al. (1995), three of the fathers being oligospermic. While it appears that the risk for chromosome abnormality in a child is small, it may nevertheless be reasonable to offer prenatal diagnosis for fathers-to-be. Seminoma. No cytogenetic abnormalities were seen in the sperm of a 26-year-old man treated with cisplatin, vinblastine, and bleomycin, analyzed some 9 months after treatment ( Jenderny et al., 1992). Hydatidiform Mole. This chromosomal condition is discussed in detail in Chapter 23. There is no increased risk for other abnormal outcome in a subsequent pregnancy, and in particular the incidence of congenital malformations is no greater (Berkowitz et al., 1994). Chemotherapy, either at the time of evacuation of complete mole, or for “persistent gestational trophoblastic tumor” following the index pregnancy, seems also to be without untoward effect in a subsequent pregnancy. Other Disease. Views differ whether there exists an increased risk for women with diabetes mellitus to have a child with Down syndrome (DS) (Narchi and Kulaylat, 1997; Martínez-Frías et al., 2002); it would be premature, as yet, to extrapolate the findings from a mouse model, in which oöcyte chromosomal misalignment was observed (Wang et al., 2009c). I N F ERT I L I TY A SS O CI AT ED W I T H CA N CER THERAPY, AND PRIOR GAMETE BANKING
Preservation of gametes prior to treatment for cancer is a logical management, and sperm banking as “fertility insurance” for boys and men with cancer Gonadal Cytogenetic Damage from Exposure
•
493
is now seen as routine (Menon et al., 2009). Blackhall et al. (2002) report 33 couples over a study period 1978–1990, the male being a survivor of Hodgkin’s, using cryopreserved sperm for artificial insemination or IVF, with nine of the 33 having 10 pregnancies. One pregnancy was terminated due to fetal hydrocephalus (46,XX karyotype); the others all produced normal infants. Cryopreservation of ovarian tissue is a more difficult procedure and still regarded as “investigational” (Tao and Del Valle, 2008). Any risk for chromosomal abnormalities in offspring is unknown, and data will need to be collected (it would be rather hypothetical to think that the risk in women might actually be reduced, if they were to reanimate their oöcytes from a younger age). I M MUN OS UP P R ESSI V E T RE AT M E NT
Azathioprine is a major immunosuppresive agent, and its inimical effect upon the ovary known (McDermott and Powell, 1996). Reports of any possible effect upon offspring are few. One example is to be noted of a dysmorphic child with two de novo abnormalities, an interstitial deletion and an apparently balanced translocation, 46,XY,del(7) (q21);t(6;14)(q21;q12), whose mother had been treated with azathioprine and prednisone for systemic lupus erythematosis, although little weight can be put on a single case report (Ostrer et al., 1984). Jenderny et al. (1992) studied a 36-year-old man who had been treated for 4 years with these same two drugs for chronic active hepatitis and showed no significant differences in sperm chromosome distributions from controls. Alkylating agents are used in the treatment of nephrotic syndrome. In women who were treated in childhood with cyclophosphamide, fertility may be little compromised, with 17 out of 18 girls in one series going on to have normal menstruation and fertility (Watson et al., 1986). Pubertal boys, and those receiving higher doses of cyclophosphamide, are likely to become azoöspermic, whereas prepubertal boys appear to have greater gonadal resistance. The use of chlorambucil for this condition is also associated with a high likelihood of azoöspermia (Levy and Stillman, 1991). OTHER PHARMACEUTICAL AGENTS
We list a few medications in which cytogenetic gonadal/gametic studies (human or animal) have been done. The reader with a particular inquiry is 494
•
NOXIOUS AGENTS
directed to the Web site of the TERIS teratogen information system, albeit that the data relate largely to teratogenic, rather than mutagenic agents, at http://depts.washington.edu/terisweb/teris/ index.html. Antimicrobials. Griseofulvin is a fungicide quite commonly used for the treatment of tinea. It is known to interfere with formation of microtubules, and it can damage the mitotic spindle. Mouse studies (although at very high dosages) suggest that meiosis may also be vulnerable, more so in oögenesis than in spermatogenesis (Shi et al., 1999). Single studies of mouse model sperm analyses suggest an increase in aneuploidies with exposure to Pyrimethamine, an antimalarial agent, and Miconazole, used in the treatment of candidiasis (Aydemir and Bilaloglu, 1996; Hassan, 1997). Whether the doses of the above agents typically used in humans might have practical reproductive implications is quite unknown. Benzodiazapines. Diazepam is used in psychiatry, and Baumgartner et al. (2001) studied patients having been on chronic high-dose treatment. Sperm disomy rates, for the chromosomes analyzed (X, Y, 13), were approximately two-fold those of controls (and sperm counts were reduced). Sex Hormones. Most studies point to no effect of previous use of the oral contraceptive pill, in terms of a subsequent risk to have a child with DS (Kallen, 1989). It is speculative whether there might be an additive effect from use of the pill together with cigarette smoking (Yang et al., 1999). Folic Acid. There was no change in prevalence of trisomy 21 in Canada comparing before and after the institution of folic acid fortification of flour in the late 1990s (Ray et al., 2003). In terms of taking periconceptual vitamin supplements, Botto et al. (2004) observed no significant difference in the frequency of the major trisomies of such pregnancies. DI AG N OST I C R A D I O LO GY
A small effect may possibly exist for DS with respect to previous X-rays to the abdomen and pelvic area; that is to say, for X-rays in which the gonads may have been within, or not far off, the field of the film. In a study of 156 mothers and 149 fathers, in whose DS children the “nondisjunctional parent” could be identified (using Q-banding polymorphisms), a
history of X-ray exposure was more often recorded in older fathers and in younger mothers (Strigini et al., 1990). The odds ratio for the whole group was 1.85, although the lower limit of the 95% confidence interval was 1.0. If such an effect truly exists in the younger mothers (and the statistics were borderline), it would seem that this slight influence becomes diluted as they get older, and the age effect comes to be predominant. NONM ED IC AL EXP OSU RES
Radioactivity. The human germline may be relatively resistant to the damaging effects of radiation, compared with some animal models (Neel et al., 1990; Adriaens et al., 2009).4 The atomic bomb blasts at Hiroshima and Nagasaki in 1945 were not followed by a statistically significant difference in the rate of chromosome abnormalities in children subsequently conceived, in a study commenced in 1967 (Awa et al., 1987; Neel and Schull, 1991; Nakamura, 2006). The study population comprised 8322 individuals born 1946–1972, age range at the time of study 12–38 years, one or both of whose parents were within 2000 meters of the hypocenter “ATB” (at the time of the bomb), alongside a contemporaneous local control group of 7976, who were either more than 2500 meters from the hypocenter or not present in the city. Sex chromosomal abnormalities were seen in 2.28 per 1000 in the former group and in 3.01 per 1000 of the latter. The only instance of autosomal trisomy was a 15-yearold with standard trisomy DS, whose father had been exposed at Hiroshima. (Given the structure of this study, deceased younger children and infants with autosomal trisomy were not included, although it is also to be noted that in separate analyses in Neel and Schull no significant correlation existed between parental exposure ATB and the frequency of stillbirth or congenital malformations.) More children of exposed parents had a small supernumerary abnormal chromosome than in the controls (5 cf. 2, a difference not specifically commented upon in Awa et al.). Of the balanced structural rearrangements, only two were confirmed as having arisen de
novo (one each in the exposed and control groups). An earlier study with specific reference to clinically diagnosed DS in 9-month-old infants, undertaken during 1948–1954 (before the chromosomal basis of DS was known), had shown no increase among offspring of 5582 exposed cf. 9452 unexposed mothers, and indeed the figures were in the other direction (0.54 cf. 1.27 per 1000), and in spite of the exposed mothers being on average slightly older (Schull and Neel, 1962). The Chernobyl nuclear plant explosion occurred in 1986, and a cloud of radioactivity was dispersed over Europe. With respect to DS, no subsequent rise in incidence was identified in a number of European countries, apart from small clusters in Berlin and Belarus, the latter of interest in that the peak was confined to 1 month, 9 months after the explosion (Little, 1993; Zatsepin et al., 2007). In contrast, Bound et al. (1995) suggest a possible link between events in 1957 (a fire in a nuclear reactor) and the early 1960s (increased levels of fallout from nuclear testing) and peaks of DS prevalence in 1958 and 1963–1964 in the Fylde district of Lancashire, England. But by no means is a firm case made: post hoc does not necessarily mean propter hoc,5 and some fluctuation is normal. The same 1957 nuclear reactor accident had been proposed as the possible reason for a cluster of six cases of DS among the children of women who had attended the same high school in Dundalk, Ireland, during 1956–1957, when they would have been aged from 12 to 19. Of the 387 births to the former pupils from this period, the expectation would have been 0.69 children with DS. However, a stringent review of the evidence, and including molecular analyses that showed one case to have arisen post fertilization, led to the conclusion that, in fact, chance was the probable basis for the “cluster” (Dean et al., 2000). While the germline, at least from the evidence outlined earlier, is apparently resistant, the same cannot be said for the bone marrow. Numerous studies on radiation exposure have shown that stable chromosome rearrangements may be induced, as measured on peripheral blood samples. Indeed, it is proposed that these changes can be used as reliable
4 Of historic interest, a very early example of ill health due to radiation exposure is that of Marie Curie, who was awarded the Nobel Prize twice. One daughter of hers was a scientist, and she also won a Nobel Prize, and the other was a skilled pianist and gifted writer. A series of n = 2 is very small, but rather evidently there must have been normal chromosomal segregation in the meioses leading to these two daughters. 5 Post hoc, ergo propter hoc (Latin) = Something happened after the event, and therefore it must have been due to the event.
Gonadal Cytogenetic Damage from Exposure
•
495
biomarkers of exposure. Populations in whom this effect has been seen include Russian nuclear plant workers, comparing those exposed to plutonium and those to gamma rays, from 1949–1989; New Zealand navy personnel who had served on ships during nuclear bomb testing in the Pacific Ocean in the late 1950s; American radiation technologists who had begun practicing from before 1950 (ages at the time of study 71–90 years); and even astronauts, unprotected by Earth’s atmosphere from solar radiation (Durante et al., 2003; Hande et al., 2003; Sigurdson et al., 2008; Wahab et al., 2008). We are unaware of any evidence that individuals exposed in these ways might have acquired any gonadal damage, and that their children could have been at risk for a chromosomal disorder. It would be a massive logistic exercise, but not without interest, if a study could be mounted of descendants of these exposed persons. Industrial Agents. Paternal occupation provides a surrogate marker for a variety of potential industrial agents. Olshan et al. (1989) assessed the father’s occupation for 1000 DS children born in British Columbia in 1952 through 1973. Seven employment categories out of 59 showed odds ratios in the range 1.4–3.3, the lower confidence limit being not less than 0.9, in certain of which exposure to various industrial agents could plausibly have occurred (including mechanics, janitors, metal workers, sawmillers). But the increases in risk were small, and with 59 items there was of course the possibility of chance fluctuation. One category that might have seemed risky, namely “other chemical workers,” in fact had the lowest odds ratio of all (0.2). Pesticides have biological activity, and it is reasonable to raise a case that distribution across the blood-testis barrier might follow inhalation, or absorption, or ingestion, and that the local effect upon gonadal tissue might be toxic. Perry (2008) has reviewed 30 studies, correlating pesticide exposure with sperm chromosomal abnormality. The methodologies varied, so direct comparisons could not readily be made. Some studies did, and some did not, show an increase in chromosomal defects. Of the numerous agents, the strongest case could be made for carbaryl and fenvalerate, in particular, as potentially causative of autosomal and gonosomal aneuploidy, with sex chromosome disomy the most frequent single abnormality. The air we breathe, it is suggested, might convey mutagens, in the form of industrial pollutants, and these might reach gonadal tissue (Somers and 496
•
NOXIOUS AGENTS
Cooper, 2009). Landfill sites contain toxic matter, which might in theory contaminate the air in nearby residential areas; but in an analysis based upon over 6000 such sites throughout the United Kingdom, and comparing populations living within, and beyond 2 kilometers of these sites, in fact no differences in the prevalence of Down syndrome were observed ( Jarup et al., 2007). However, older mothers living within one mile of industrial sites from which solvent and heavy metal emissions are vented may have a slightly increased risk for aneuploidies in their offspring (Brender et al., 2008); in the male, variation in air pollution may affect some aspects of sperm quality, although with no obvious influence upon disomy or diploidy rates (Rubes et al., 2005). Confirmatory studies are needed. Bisphenol A. Bisphenol A (BPA), an estrogenic chemical used widely in plastic manufacture, has been shown to disrupt several different stages of oöcyte development in mice (Susiarjo et al., 2007). The fetal ovaries observed after pregnant mice were treated with low, environmentally relevant doses of BPA during mid-gestation showed synaptic defects and increased levels of recombination. The mature females, exposed as fetuses, went on to have oöcytes and embryos with aneuploidies. There may be further environmental influences on the effect of BPA in that variations in diet influenced the observation of meiotic abnormalities in exposed mice (Muhlhauser et al., 2009), thus demonstrating the complexities in studying environmental exposures, since even in laboratory animals it is nearly impossible to keep all other variables constant. Reservations are already held concerning its use in human activity for other health-related reasons, and the mouse data might be seen as one further reason for caution. Agent Orange. Agent Orange (a mixture of phenoxylic herbicides) was used in the Vietnam War as a defoliant spray, and those exposed may have absorbed the chemical via the oral route in particular. A study of New Zealand Vietnam veterans some 3–4 decades after the war showed an increase in sister chromatid exchanges on blood samples (Rowland et al., 2007). Whether gonadal genetic damage results is controversial (Ngo et al., 2006; Schecter and Constable, 2006; Fraser, 2009); specifically, we are unaware of any evidence for an increased risk of chromosomal abnormalities in offspring. Tobacco, Alcohol, Caffeine. Tobacco smoking in mothers had no influence upon the incidence of DS in the study of Chen et al. (1999a), based upon data of a population case-control analysis in Washington
state from 1984–1994, and in which they had been at pains to account for a confounding effect of maternal age. The odds ratio was exactly 1.0—that is, no effect either way—for smokers versus nonsmokers. Similar findings are also reported from Sweden, California, and England (Kallen, 1997; Torfs and Christianson, 2000; Rudnicka et al., 2002). Nevertheless, a tentative role has been proposed for one particular mechanism: trisomy 21 due to nondisjunction in maternal meiosis II (MMII). In a case-control study in Atlanta, cigarette smoking around the time of conception gave an odds ratio of 7.6 in mothers of MMII trisomic offspring, compared with controls, in the <35-year age group (Yang et al., 1999). Very speculatively, smoking might diminish blood flow in the microvasculature of the perifollicular bed, and the resultant hypoxia could compromise some aspect of the oöcyte’s functioning as the meiotic process is reactivated. Alcohol and coffee taken by the mother prior to conceiving might actually reduce the DS risk. In the study of Torfs and Christianson, the odds ratios for “high” alcohol and coffee consumption (≥4 drinks per week, ≥4 cups per day) were 0.54 and 0.63, respectively. If these figures reflect biological reality, a possible mechanism would be a selective reduction in viability of a trisomic 21, as compared to a normal conceptus. Concerning spermatogenesis, Shi and Martin (2000b), reviewing the literature, concluded that personal habits with respect to smoking, alcohol, and caffeine ingestion appear not to have any consistent effect upon disomy rates in sperm, although since there were somewhat varying findings in the different studies, it had to be acknowledged that a definitive answer was not at hand. Shi et al. (2001a) proceeded to a study of cigarette smoking and aneuploidy using FISH analysis of sperm, with reference to chromosomes 13, 21, X and Y. They divided their subjects into nonsmokers, light smokers (<20 cigarettes/day), and heavy smokers (≥20/day). The smokers showed an increase in disomic 13 sperm (0.2% of sperm 24,+13, versus 0.07% in controls), which was statistically significant. The rates of disomies 21, X and Y were within the control ranges. Chromosome 13 and, from other studies, chromosome 1 may be more susceptible, as they go through meiotic disjunction, to an untoward influence of toxic substances in cigarette smoke. Since most trisomy 13 is due to a maternal meiotic error, and given
that the excess is, in absolute terms, very small, it seems safe to suppose that fathers who smoke contribute scarcely, if at all, to the totality of this particular aneuploidy. As for alcohol, the observation of a negative association between sperm disomy frequencies and alcohol consumption in one study6 (Härkönen et al., 1999), and noting also the figures earlier on maternal consumption, should not lead one to advise that couples drink more heavily prior to a planned impregnation! Second-hand smoke is difficult to assess, outside of controlled animal studies (Hung et al., 2009). A study of four adult male rhesus macaques, exposed to second-hand smoke for 6 months, showed no change in the X:Y ratio in sperm, which may indicate that there is no increase in aneuploidy. In addition, second-hand smoke-exposed pregnancies did not show increased DNA damage in their offspring, as compared to babies born to nonsmoking mothers (de Assis et al., 2009).
GENETIC COUNSELING As Wyrobek and Adler (1996) commented, “it has been more than half a century [1927] since Muller demonstrated that X-rays can induce germinal mutations in Drosophila, yet questions as basic as the existence of even a single human germinal mutagen remain unresolved.” McFadden and Friedman, writing in 1997, noted that no environmental agent has been identified in which it could be stated, beyond reasonable doubt, that this agent would cause chromosome abnormalities in the offspring of exposed parents. While some studies have shown increased rates of aneuploidy in sperm, the practical fact remains that there is no excess in children born with chromosomal syndromes. Only in 2001 could Marchetti et al. claim, with respect to their work on etoposide exposure with a mouse model, that “we know of no other report of an agent for which paternal exposure leads to an increased incidence of aneuploidy in the offspring.” Encouragement can be drawn from this largely negative information, and the counselor will usually be justified in offering substantially reassuring advice from the particular focus of chromosome abnormality. Reference to the commentaries earlier may provide useful supporting information for the individual agent of specific interest. Prenatal diagnosis would
6 But another study showed a positive association with alcohol, as well as with caffeine (Robbins et al., 1997).
Gonadal Cytogenetic Damage from Exposure
•
497
be a discretionary option, as would be preimplantation diagnosis for those whose treatment-related infertility required IVF. A propos the specific question of previous treatment for cancer, Byrne (1999) does remain cautious, considering that on the rather few data available there are only “limited grounds for reassurance,” and seeing us as being “in the infancy of studies of germ cell mutagenesis in cancer
498
•
NOXIOUS AGENTS
survivors.” She emphasizes that the opportunity to assess newer cancer treatments has not yet arisen, and that the timeframe for these assessments is to be measured in decades. Certainly, and more particularly for those agents in which a biological link could plausibly be proposed, it is right that research in this area continue.
appendixes appendix a: ideograms of human chromosomes, and haploid autosomal lengths
HAP LO ID AUTOS O M A L LE N GT H
To determine the (quantitative) amount of a particular segmental imbalance, as a fraction of the haploid autosomal length (HAL), multiply (1) the fraction of the whole chromosome that this segment comprises by (2) the haploid autosomal length (HAL) of the whole chromosome. The fraction is readily estimated by placing a millimeter rule against the ideogram (Fig. A–1) (or by reference to the relative distances between borders of bands listed in Table 1 in Francke, 1994). The HAL of the autosome concerned is taken from Table A–1.
For example, considering the imbalance due to the karyotype of the children pictured in the frontispiece and shown in Figure 4–1, what proportion of the HAL does the segment 4q31.3→qter constitute? First, the segment comprises 18% of the length of chromosome 4: running a millimeter rule alongside the ideogram of chromosome 4 in Figure A–1, the whole chromosome is 116 mm and the segment is 21 mm, and 21/116 = 18%. Second, from the table, chromosome 4 is 6.30% of the total HAL. Thus, 18% of 6.30% = 1.15% of HAL.
•
499
Table A–1. The Percentage of Haploid Autosomal Length (HAL) That Each Autosome Constitutes CHROMOSOME
PERCENTAGE OF HAL
CHROMOSOME
PERCENTAGE OF HAL
9.24 8.02 6.83 6.30 6.08 5.90 5.36 4.93 4.80 4.59 4.61
12 13 14 15 16 17 18 19 20 21 22
4.66 3.74 3.56 3.46 3.36 3.46 2.93 2.67 2.56 1.90 2.04
1 2 3 4 5 6 7 8 9 10 11 Source: From Daniel (1985).
500
•
APPENDIXES
36.33 36.31 36.32 36.23 36.22 36.21 36.13 36.12 36.11
25.3 25.2 25.1
35.3 35.2 35.1 34.3 34.2 34.1
24.3 24.2 24.1 23.2
33 32.3 32.2
21.2
23.3 23.1 22.3 22.2 22.1 21.3
32.1
21.1
31.3 31.2
16.3 16.2 16.1
31.1
26.2
15 22.3 22.2
13.3
14 13.2
22.1 13.1 21.3 21.2 21.1
11.2 11.1 11.1 11.2
13.2 13.1 12 11.2 11.1 11
14 13 12 11 11
23.2 23.3 24.1 24.2 24.3 25.1 25.2
24.3
12.2 12.3 13.11 13.12 13.13 13.2 13.31 13.32 13.33 21.2 21.1
31.1
21.3
27
22.1 22.3
28.1
23
28.3
22.2
31.2
31.1 31.2 31.3
31.3 32.1
32.11 32.12 32.13 32.2 32.3 41 42.11 42.12
33.1 33.2 33.3
36.2
1
31.1 31.21 31.22 31.23 31.3 32.1 32.2 32.3 33 34.1 34.2 34.3
26.2 26.31 26.32 26.33 27.1 27.2 27.3 28
37.1 37.2 37.3
44
28.2
26.1
34 35 36.1 36.3
42.2 42.3 43
26
24 25.1 25.2 25.32 25.31 25.33
32.2 32.3
42.13
21.1 21.21 21.22 21.23 21.3 22.1 22.2 22.3 23 24 25
12.1
24.2
25.3
13.1 13.2 13.3
12.3 12.2 12.1 11.2 11.1 11.1 11.2
22.1 22.2 22.3 23.1 23.2 23.3 24.1
23.1
12
13
21.2 21.3
21.2 21.3 22
15.1
21.2 21.1 14.3 14.2 14.1
21.1 21.1
15.2
21.31
12.212.1 12.3 13 14.1 14.2 14.3
12
16.3 16.2 16.1 15.33 15.32 15.31
22.2 22.1 21.33 21.32
12
13.3
26.3 26.1 25.3 25.2 25.1 24.3 24.2 24.1 23 22.3
35.1
29
2
35.2
3
4
FIGURE A–1 These depictions represent the bands that can be distinguished at a very high level (850 band) of cytogenetic resolution. Different shadings and lengths of bands reflect actual intensities and lengths as observed by the cytogeneticist. (From N.L. Chia, 2009, A comprehensive set of idiograms representing all interpretive levels of resolution: ISCN (2009), Cytogenet Genome Res 125: 162–164. © 2009 Nicole Chia)
15.33 15.32 15.31 15.2
25.3
25.2 25.1 24 23 22.33 22.32 22.31 22.2 22.1 21.33 21.32 21.31 21.2
15.1 14.3 14.2 14.1 13.3 13.2 13.1 12 11 11.1
12 13 14.2
21.3 21.1 15.3 15.2
23.3 23.2 23.1
15.1 14.3 14.2 14.1 13 12.3 12.2 12.1 11.2 11.1 11.1 11.21
12.3 12.2 11.2 12.1 11.1 11.1 11.2
14.1 14.2 14.3
22 21.3 21.2 21.1 12 11.23 11.22 11.21 11.1 11.1 11.21
14.1 14.3
11.22
11.22
11.23
15
11.23 12.2 12.1 12.3 13.2 13.1 13.3 21.11 21.12 21.13 21.2 21.3
15
21.1 21.2 21.3 22.1
21.11
16.1 16.2 16.3 21.1 21.2 21.3
22.3 23.1 23.2 23.3
21.12 21.13 21.2 21.3 22.1
22.1 22.2 22.31 22.32 22.33 23.1 23.2
31.1 31.2 31.3
22.3
25.1
15.3 15.2 15.1 14
22.3 22.2 22.1 21.3
12.32
21.1 13.3 13.2 13.1 12 11.2 11.1 11
13 12.33 12.31 12.2 12.1
14.3
11.2 11.12 11.11 11 12.1 12.2 12.3
21.1 21.2 21.3
13.1
22.1 22.2 22.3 23.1 23.2 23.31 23.32 23.33 24.2 24.1 24.31 24.32 24.33 25.1 25.2 25.3 26.11 26.12
22.1 22.2 22.31 22.32 22.33 31.1 31.2 31.3 32 33.1 33.2 33.3 34.1 34.2
13.2 13.3 13.4 13.5
FIGURE A–1
(Continued)
21.33
24.11 24.12 24.13 24.21 24.22 24.23
23.3 24.1 24.2 24.3 25
10
13.11 13.12 13.13 13.2 13.3 14.1 14.2 14.3 15 21.1 21.2 21.31
22 23.1 23.2 23.3
21 22.1 22.2 22.3 23.1 23.2
26.2 26.3
9
21.32
14.1 14.2 14.3
26.13
34.3
8 13.33 13.32 13.31 13.2 13.1 12.3 12.2 12.1 11.23 11.22 11.21 11.1 11 12
14.2 14.1 13 12
11.22 11.21 11.1 11.1 11.21 11.22 11.23
13 21.11 21.12 21.13 21.2 21.31 21.32 21.33
24.3
15.5 15.4 15.3 15.2 15.1
11.23
12
24.21 24.22 24.23
7
6
24.3 24.2 24.1 23
22.3 23.1 23.2 23.3 24.11 24.12 24.13
35 36.1 36.2 36.3
25.3 26 27
5
22.1 22.2
34
25.2
34 35.1 35.2 35.3
22.2
31.1 31.2 31.31 31.32 31.33 32.1 32.2 32.3 33
23.3 24.1 24.2 24.3
32 33.1 33.2 33.3
21.2
21.2
21.1
11.2 12.1 12.2 12.3 13.1 13.21 13.22 13.23 13.3
22.2
22.3 22.2 22.1
24.31 24.32 24.33
11
12
13 12
13 12 11.2 11.1 11.1
11.2 11.1 11 12.11 12.12 12.2
12.13
11.2
12.3 13.1
13.2
12
13.3 14.11 14.12 14.13 14.2 14.3
21.32
13.3
21.1 21.2 21.31
23.2
13.1 13.2 21.1 21.2 21.3
13.2
23.3 24.1
31.1 31.2 31.3 32.1 32.2
24.3
34
13
11.2 11.1 11.1 11.2 12.1 12.2 13
24.1 24.2 24.3 25.1 25.2 25.3
31.1 31.2 31.3 32.11 32.12 32.13 32.2 32.31 32.32 32.33
33.3
13.2 13.13 13.12 13.11 12.3 12.2 12.1
14 15.1 15.2 15.3 21.1 21.2 21.3 22.1 22.2 22.31 22.32 22.33 23
24.2
32.3 33.1
13.3
11.2 11.1 11.1 11.2 12 13.1 13.3
22.1 22.2 22.3 23.1
21.33 22.1 22.2 22.3
33.2
13 12
21 22.1 22.2 22.3 23.1 23.2 23.3
26.1 26.2 26.3
14
24.1 24.2 24.3
16
15
13.3 13.2 13.1
11.32 11.31 11.23 11.22 11.21
12 11.2 11.1 11.1
13.3
11.1 11.1
11.2 12 21.1 21.2
11.2
21.31
12.1 12.2 12.3
21.32
21.1
21.33 22 23.1 23.2 23.3 24.1 24.2 24.3 25.1 25.2 25.3
21.2 21.31 21.32 21.33 22.1 22.2 22.3 23
17 FIGURE A–1
13.11
11.23 11.22
12 11 11 12 13.11 13.12 13.13 13.2 13.31 13.32
11.21 11.1 11.1 11.21 11.22 11.23 12 13.11 13.12 13.13
13.33
13.2 13.31 13.32 13.33
13.41 13.42 13.43
18 (Continued)
13 12.3 12.2 12.1
13.2 13.13 13.12
19
20
13 12 13 12 11.2
11.2
11.1 11.1
11.21
22.33 22.32 22.31 22.2
11.1 11.1
11.2 21.1
22.13 22.12 22.11 21.3 21.2 21.1
11.22 11.23 12.1
21.2 21.3 22.11 22.12 22.13 22.2 22.3
12.2
12.3 11.4 11.3 11.23
13.1 13.2 13.31 13.32 13.33
11.22 11.21 11.1 11.1
21
22
11.2 12 13.1 13.2 13.3 21.1 21.2 21.31 21.32 21.33 22.1
11.32 11.31
22.2 22.3
11.2
23
11.1 11.1
24
11.21 11.221 11.222 11.223 11.23
25 26.1 26.2 26.3 27.1 27.2 27.3
12
28
Y FIGURE A–1
(Continued)
X
appendix b: cytogenetic abbreviations and nomenclature
CYTOGENETICS HAS its own jargon and fondness for acronyms, and certain abbreviations are regularly used. The ICSN (itself an abbreviation for International System of Cytogenetic Nomenclature; ISCN, 2009) provides formally approved abbreviations, most of which are set out next; and following this, we list another set of abbreviations (and including a number of clinical expressions), which are used fairly frequently in this book and in many genetics journals, and which should be familiar to the reader:
SOME ISCN ABBREVIATIONS add cht del der dic
Additional material of unknown origin Chromatid Deletion Derivative chromosome Dicentric chromosome
dim
Deletion, identified by CGH (= diminished) dn De novo dup Duplication enh Duplication, identified by CGH (= enhanced) fis Fission (at the centromere) fra Fragile site h Secondary constriction i Isochromosome idem the same (to avoid repetition of complex description in a mosaic case) ins Insertion dir ins Direct insertion inv ins Inverted insertion ish In situ hybridization inv Inversion mar Marker chromosome mat Maternal origin min Minute chromosome minus (–) Loss of a whole chromosome •
505
mos p pat plus (+) q r rcp rea rec rob solidus (/) stk t tan tas ter upd
Mosaic Short arm Paternal origin Gain of a whole chromosome Long arm Ring Reciprocal translocation Rearrangement Recombinant chromosome Robertsonian translocation Separates cell lines in describing mosaics Satellite stalk Translocation Tandem Telomere association Terminal (end of chromosome arm) Uniparental disomy
SOME OTHER COMMONLY USED ABBREVIATIONS abn(X) array-CGH AFP arr AS AT BAC CGH CpG CPM CT CV CVS DGS DMR DS ECARUCA
ESAC
506
•
An abnormal X chromosome Microarray comparative genomic hybridization Alpha-fetoprotein Array (microarray) Angelman syndrome Ataxia-telangiectasia Bacterial artificial chromosome Comparative genomic hybridization Cytosine-guanine DNA sequence Confined placental mosaicism Computed tomography (an organ imaging modality) Chorionic villus Chorionic villus sampling DiGeorge syndrome Differentially methylated region Down syndrome European Cytogeneticists Association Register of Unbalanced Chromosome Aberrations Extra structurally abnormal chromosome
APPENDIXES
ESHRE
European Society for Human Reproduction and Embryology EUCROMIC European collaborative research group on mosaicism in CVS FA Fanconi anemia FISH Fluorescence in situ hybridization FRAXA Fragile XA syndrome FSH Follicle stimulating hormone HAL Haploid autosomal length ICSI Intracytoplasmic sperm injection IUGR Intrauterine growth retardation IVF In vitro fertilization Kb Kilobases of DNA KS Klinefelter syndrome LCRs Low copy repeats Mb Megabases of DNA MCA/MR Multiple congenital anomalies/ mental retardation MLPA Multiplex ligation-dependant probe analysis MLS Microöphthalmia with linear skin defects MRI Magnetic resonance imaging (an organ imaging modality) mtDNA Mitochondrial DNA NAHR Nonallelic homologous recombination NHEJ Nonhomologous end joining NOR Nucleolar organizing region nt Nucleotide OMIM Online mendelian inheritance in man PAR (1 and 2) Pseudoautosomal region (primary and secondary) PB (1 and 2) Polar body (first and second) PCD Premature centromere division PCR Polymerase chain reaction PGD Preimplantation diagnosis PGD-AS Preimplantation diagnosis for aneuploidy screening PND Prenatal diagnosis POF Premature ovarian failure PWS Prader-Willi syndrome PWACR Prader-Willi/Angelman critical region RIF Recurrent implantation failure RP11 Roswell Park BAC clone library 11 RPL Recurrent pregnancy loss RT-PCR Real-time polymerase chain reaction SCE Sister chromatid exchange
SMC snoRNA SNP TS UCSC UPD UTR WHS
Supernumerary marker chromosome Small nucleolar RNA Single nucleotide polymorphism Turner syndrome University of California at Santa Cruz (hosts a Genome Browser) Uniparental disomy Untranslated region Wolf-Hirschhorn syndrome
46,XY,Yqh–
Normal male, deletion of material from heterochromatic region of Y long arm 46,XY,fra(10) Normal male, fragile site on (q23.3) chromosome 10 long arm at subband 23.3
A BN O R M A L
Sex Chromosome Aneuploidies. KARYOTY P E N O MENC LAT U RE ACCORDING TO THE ISCN
The description of chromosomal constitution in most laboratory reports and in most case reports in the literature is the ISCN (2009) format. First, the diploid number is given. Second, the sex chromosome constitution is given. Thereafter, any abnormality or variant is described. Certain abbreviations are used, as listed earlier. In structural rearrangements, the position of breakpoints is given by reference to the band involved: short or long arm (p or q), region, and band or subband(s) within that band. The region is denoted by a digit 1 through 4, the band by a digit 1 through 8, and the subband(s) by digit(s) following a “decimal point.” The centromere is p10 or q10. Illustrative examples of commonly described karyotypes follow. The nomenclature has evolved to accommodate the growing complexity of cytogenetics, with the earlier nomenclatures issued since the first in 1960, often referred to by the name of the city in which the committee met (Denver, London, Chicago, Paris), and subsequently more anonymously as ISCN (year). Published papers from the earlier years will, of course, have used the nomenclature of their time. Many old papers remain a valuable resource, particularly case reports. The reader consulting these may therefore need to adjust and learn to handle earlier (generally simpler) versions of cytogenetic nomenclature.
45,X
Monosomy X (Turner syndrome) Klinefelter syndrome Triple X female XYY “syndrome” Two of the more common types of polysomy X Mosaic Klinefelter syndrome*
47,XXY 47,XXX 47,XYY 48,XXXX 49,XXXXY 47,XXY/46,XY or mos 47,XXY/46,XY 45,X/46,XX Mosaic Turner syndrome*
Autosomal Aneuploidies.
47,XY,+21 47,XX,+21/46,XX 47,XX,+18 47,XY,+13 47,XX,+8/46,XX 47,XY,+16 45,XX,–21
Trisomy 21 (Down syndrome) Mosaic Down syndrome* Trisomy 18 (Edwards syndrome) Trisomy 13 (Patau syndrome) Mosaic trisomy 8* Trisomy 16 Monosomy 21
Polyploidies.
EXAMPLES OF CYTOGENETIC NOMENCLATURE 46,XX 46,XY 46,XX,9qh+
69,XXY 92,XXXX
Triploidy Tetraploidy
Normal female Normal male Normal female, additional material in heterochromatic region of chromosome 9 long arm
APPENDIXES
•
507
Deletions. 46,XX,del(4)(p15)
Terminal deletion chromosome 4 short arm (Wolf-Hirschhorn syndrome) 46,XX,del(5)(p13) Terminal deletion chromosome 5 short arm (cri du chat syndrome) 46,XX,del(18)(q12) Terminal deletion chromosome 18 long arm Reciprocal Translocations 46,XX,t(4;12) Reciprocal translocation (p14;p13) between chromosome 4 and 12, with breakpoints at p14 in chromosome 4 and p13 in chromosome 12 46,XY,der(12)t(4;12) Unbalanced complement, (p14;p13)mat having received derivative chromosome 12 in place of normal 12 from translocation carrier mother 47,XX,+der(22) Unbalanced complement t(11;22)(q23;q11) having received pat derivative 22 as a supernumerary chromosome from translocation carrier father
508
•
APPENDIXES
Fish Example: 46,der(15)t(8;15) Unbalanced complement, (q22.3;q26.2) having received the mat.ish derivative 15 in place of a normal 15 from the carrier der(15)t(8;15) mother, resulting in partial (qter+;qter-) deletion of 15q and an extra copy of the chromosome 8 segment Microarray Example: 46,der(15)t(8;15) Unbalanced complement, (q22.3;q26.2) having received the mat, arr derivative 15 in place of 8q22.3q24.3 normal 15 from the carrier (105,171,556– mother. On microarray, 146,201,91)×3, the extra segment 15q26.2q26.3 extends from nucleotides (96,062,102– 105,171,556 to 146,201,91 100,201,136)×1 on chromosome 8, and the deleted segment from 96,062,102 to 100,201,136 on chromosome 15.
WHOLE- ARM R EC IP ROC A L TRANSLO C ATIO N S
46, t(1;9) Balanced carrier of translocation (p10;p10) having both breakpoints at the centromeres, with exchange of whole short arms. Translocation chromosomes are 9p/1q and 1p/9q 46,t(3;4) Balanced carrier of translocation (p10;q10) having both breakpoints at the centromeres, with exchange of whole short arm of chromosome 3 and whole long arm of chromosome 4. Translocation chromosomes are 3q/4q and 3p/4p Robertsonian Translocations. 45,XY,der(14;21) (q10;q10) (or replace der with rob)
Balanced carrier of monocentric fusion Robertsonian translocation between chromosomes 14 and 21 46,XY,der(14;21) Unbalanced complement, (q10;q10)mat, having received +21 (or replace der (14;21) Robertsonian with rob) chromosome as well as a “free” chromosome 21 from mother (the karyotype of translocation Down syndrome) Inversions. 46,XX,inv(3) (p23q27)
Inversion (pericentric) of chromosome 3, breakpoints at p23 and q27 46,XY,rec(3) Recombinant chromosome has dup(3p) been transmitted from mother inv(3) carrying inversion chromosome (p23q27)mat 3. There is duplication of the short arm segment distal to p23; and deletion of the long arm segment distal to q27 46,XY,inv(11) Inversion (paracentric) of (q13q22) chromosome 11, breakpoints at q13 and q22
Insertions.
46,XY,dir Direct insertion of segment ins(10;8) q21.2→q22 of chromosome (q21;q21.2q22) 8 into q21 of chromosome 10. Segment has original orientation to centromere, namely, q21.2 is proximal and q22 distal 46,XX,inv ins(2) Inverted insertion of segment (p13q31q21) q31→q21 into band p13. Segment has opposite orientation to centromere, namely, q31 is proximal and q21 distal Other. 46,XY,t(1;18;15) (q32;q21;q24)dn
De novo complex translocation, involving exchanges between three chromosomes, at the breakpoints indicated 46,XX,r(15) A ring 15 chromosome 46,X,i(Xq) An isochromosome of the X long arm 46,XX,add(19)(p13) Additional material of unknown origin attached to band p13 of chromosome 19 46,XY,upd(15)mat Uniparental disomy for a maternally derived chromosome 15 46,Y,fra(X)(q27.3) Male with a fragile site in subband 27.3 on the X long arm 1(pp)(qqqqqqqqqq) Multiradial of chromosome 1 comprising two short arms and ten long arms (informal nomenclature) (and see Fig. 19–4) *In mosaicism, the normal cell line is listed last.
APPENDIXES
•
509
appendix c: determining 95 percent confidence limits, and the standard error
Confidence Limits The “Exact confidence limits for p” tables in Documenta Geigy (1982, pp. 89–102) are a useful source of data on confidence limits for the sizes of sample geneticists generally collect. Suppose in a kindred—ascertainment bias having been suitably accounted for—of a total of 54 offspring of translocation carriers, 5 were abnormal, and 49 were phenotypically normal. The frequency for abnormality from this particular sample is 9.3% (5/54). Checking in Documenta Geigy under N = 54, x = 5, we see that the 95% confidence limits are given as 3.08% to 20.30%. In other words, we may take it as close to being sure that the true risk lies in the range 3% to 20%. Calculation The standard error (SE) is calculated from the simple formula
510
•
SE =
a (n − a ) n3
where a = the number of abnormals, and n = the total number of offspring after ascertainment correction (Stengel-Rutkowski et al., 1988). Thus, for the preceding example 5 (54 − 5) 54 3 = 0.039
SE =
And thus the risk is given as 9.3 ± 3.9%.
references
Aalfs CM, Fantes JA , Wenniger-Prick LJJM, et al. Tandem duplication of 11p12-p13 in a child with borderline development delay and eye abnormalities: dose effect of the PAX6 gene product? Am J Med Genet 73: 267–271, 1997. Abaied L, Trabelsi M, Chaabouni M, et al. A novel UBE3A truncating mutation in large Tunisian Angelman syndrome pedigree. Am J Med Genet 152A : 141–146, 2010. Abeliovich D, Dagan J, Lerer I, Silberstein S, Katznelson MB, Frydman M. t(15;21)(q15;q22.1) pat resulting in partial trisomy and partial monosomy of chromosomes 15 and 21 in two offspring. Am J Med Genet 66: 45–51, 1996. Abeliovich D, Yagupsky P, Bashan N. 3:1 meiotic disjunction in a mother with a balanced translocation, 46,XX,t(5,14)(p15;q13) resulting in tertiary trisomy and tertiary monosomy offspring. Am J Med Genet 12: 83–89, 1982. Aboura A , Labrune P, Perreaux F, et al. Deletion in the ABL gene resulting from a meiotic recombination of a maternal (3;22;9)(q22;q12;q34.1) translocation. J Med Genet 40: e6, 2003.
Abrams DJ, Aronoff AR , Berend SA , Roa BB, Shaffer LG, Geier MR . Prenatal diagnosis of a homologous Robertsonian translocation involving chromosome 15. Prenat Diagn 21: 676–679, 2001. Abrams DJ, Augustyn AM, Geier MR . Prenatally diagnosed mosaic trisomy 17: a case report with two-year follow-up. Prenat Diagn 25: 968–969, 2005. Abrams L, Cotter PD. Prenatal diagnosis of de novo X;autosome translocations. Clin Genet 65: 423–428, 2004. Abramsky L, Chapple J. Prenatal Diagnosis: The Human Side. 2nd ed. Cheltenham, England: Nelson Thornes, 2004. Abramsky L, Hall S, Levitan J, Marteau TM. What parents are told after prenatal diagnosis of a sex chromosome abnormality: interview and questionnaire study. Br Med J 322: 463–466, 2001. Abuelo DN, Barsel-Bowers G, Richardson A . Insertional translocations: report of two new families and review of the literature. Am J Med Genet 31: 319–329, 1988.
•
511
Adachi M, Tachibana K , Asakura Y, Muroya K , Ogata T. Del(X)(p21.1) in a mother and two daughters: genotype-phenotype correlation of Turner features. Hum Genet 106: 306–310, 2000. Adeyinka A , Adams SA , Lorentz CP, Van Dyke DL, Jalal SM. Subtelomere deletions and translocations are frequently familial. Am J Med Genet 135: 28–35, 2005. Adeyokunnu AA . The incidence of Down’s syndrome in Nigeria. J Med Genet 19: 277–279, 1982. Adhvaryu SG, Peters-Brown T, Livingston E, Qumsiyeh MB. Familial supernumerary marker chromosome evolution through three generations. Prenat Diagn 18: 178–181, 1998. Adriaens I, Smitz J, Jacquet P. The current knowledge on radiosensitivity of ovarian follicle development stages. Hum Reprod Update 15: 359–377, 2009. Aiello V, Ricci N, Palazzi P, D’Agostino G, Azzini G, Calzolari E. New variant of chromosome 11. Am J Med Genet 50: 294–295, 1994. Aisner J, Wiernik PH, Pearl P. Pregnancy outcome in patients treated for Hodgkin’s disease. J Clin Oncol 11: 507–512, 1993. Aït Yahya-Graison E, Aubert J, Dauphinot L, et al. Classification of human chromosome 21 geneexpression variations in Down syndrome: impact on disease phenotypes. Am J Hum Genet 81: 475–491, 2007. Akahoshi K , Fukai K , Kato A , Kimiya S, Kubota T, Spritz RA . Duplication of 15q11.2-q14, including the P gene, in a woman with generalized skin hyperpigmentation. Am J Med Genet 104: 299–302, 2001. Akahoshi K , Spritz RA , Fukai K , Mitsui N, Matsushima K , Ohashi H. Mosaic supernumerary inv dup(15) chromosome with four copies of the P gene in a boy with pigmentary dysplasia. Am J Med Genet 126A : 290–292, 2004. Akcakus M, Ozkul Y, Gunes T, et al. Associated anomalies in asymmetric crying facies and 22q11 deletion. Genet Couns 14: 325–330, 2003. Albrecht B, Mergenthaler S, Eggermann K , Zerres K , Passarge E, Eggermann T. Uniparental isodisomy for paternal 2p and maternal 2q in a phenotypically normal female with two isochromosomes, i(2p) and i(2q). J Med Genet 38: 214, 2001. Albrecht U, Sutcliffe JS, Cattanach BM, et al. Imprinted expression of the murine Angelman syndrome gene, Ube3a, in hippocampal and Purkinje neurons. Nat Genet 17: 75–78, 1997. Aldhous MC, Abu Bakar S, Prescott NJ, et al. Measurement methods and accuracy in copy number variation: failure to replicate associations of beta-defensin copy number with Crohn’s disease. Hum Mol Genet 15: 4930–4938, 2010.
512
•
REFERENCES
Aldred MA , Aftimos S, Hall C, et al. Constitutional deletion of chromosome 20q in two patients affected with albright hereditary osteodystrophy. Am J Med Genet 113: 167–172, 2002. Aleck KA , Argueso L, Stone J, Hackel JG, Erickson RP. True hermaphroditism with partial duplication of chromosome 22 and without SRY. Am J Med Genet 85: 2–4, 1999. Alfarawati S, Fragouli E, Colls P, Wells D. First births after preimplantation genetic diagnosis of structural chromosome abnormalities using comparative genomic hybridization and microarray analysis. Hum Reprod 26: 1560–1574, 2011. Algar EM, St Heaps L, Darmanian A , et al. Paternally inherited submicroscopic duplication at 11p15.5 implicates insulin-like growth factor II in overgrowth and Wilms’ tumorigenesis. Cancer Res 67: 2360–2365, 2007. Al-Kouatly HB, Chasen ST, Gilbert F, Ahner R , Alonso LM, Chervenak FA . Correlation between rare chromosomal abnormalities and prenatal ultrasound findings. Am J Med Genet 107: 197–200, 2002. Allanson JE, Greenberg F, Smith AC. The face of Smith-Magenis syndrome: a subjective and objective study. J Med Genet 36: 394–397, 1999. Allderdice PW, Ali M, McAlpine PJ. Complementation by two non-homologous recombinant chromosomes 3. Am J Med Genet 39: 396–398, 1991. Allderdice PW, Browne N, Murphy DP. Chromosome 3 duplication q21 leads to qter deletion p25 leads to pter syndrome in children of carriers of a pericentric inversion inv(3) (p25q21). Am J Hum Genet 27: 699–718, 1975. Allderdice PW, Eales B, Onyett H, et al. Duplication 9q34 syndrome. Am J Hum Genet 35: 1005–1019, 1983. Allen EG, Freeman SB, Druschel C, et al. Maternal age and risk for trisomy 21 assessed by the origin of chromosome nondisjunction: a report from the Atlanta and National Down Syndrome Projects. Hum Genet 125: 41–52, 2009. Allingham-Hawkins DJ, Babul-Hirji R , Chitayat D, et al. Fragile X premutation is a significant risk factor for premature ovarian failure: the International Collaborative POF in Fragile X study—preliminary data. Am J Med Genet 83: 322–325, 1999. Alter BP, Rosenberg PS, Brody LC. Clinical and molecular features associated with biallelic mutations in FANCD1/BRCA2. J Med Genet 44: 1–9, 2007.
Alvarez-Nava F, Gonzalez S, Soto M, Martinez C, Prieto M. Complete androgen insensitivity syndrome: clinical and anatomopathological findings in 23 patients. Genet Couns 8: 7–12, 1997. Alves C, Carvalho F, Cremades N, Sousa M, Barros A . Unique (Y;13) translocation in a male with oligozoospermia: cytogenetic and molecular studies. Eur J Hum Genet 10: 467–474, 2002. Amati F, Conti E, Novelli A , et al. Atypical deletions suggest five 22q11.2 critical regions related to the DiGeorge/velo-cardio-facial syndrome. Eur J Hum Genet 7: 903–909, 1999. American Academy of Pediatrics Committee on Bioethics. Sterilization of women who are mentally handicapped. Pediatrics 85: 868–871, 1990. American College of Obstetricians and Gynecologists Committee on Genetics. ACOG committee opinion. No. 338: screening for fragile X syndrome. Obstet Gynecol 107: 1483–1485, 2006. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 77: screening for fetal chromosomal abnormalities. Obstet Gynecol 109: 217–227, 2007. American College of Obstetricians and Gynecologists Committee on Genetics. ACOG Committee Opinion No. 446: array comparative genomic hybridization in prenatal diagnosis. Obstet Gynecol 114: 1161–1163, 2009. American Society of Human Genetics Board of Directors & American College of Medical Genetics Board of Directors. Points to consider: ethical, legal, and psychosocial implications of genetic testing in children and adolescents Am J Hum Genet 57: 1233–1241, 1995. American Society for Reproductive Medicine, Practice Committee. Definitions of infertility and recurrent pregnancy loss. Fertil Steril 90: S60, 2008. Amor DJ, Bentley K , Ryan J, et al. Human centromere repositioning “in progress.” Proc Natl Acad Sci USA 101: 6542–6547, 2004. Amor DJ, Cameron C. PGD gender selection for non-Mendelian disorders with unequal sex incidence. Hum Reprod 23: 729–734, 2008. Amor DJ, Choo KHA . Neocentromeres: role in human disease, evolution, and centromere study. Am J Hum Genet 71: 695–714, 2002. Amor DJ, De Crespigny L, Gardner RJM. Urinary tract defects and chromosomal disorders. In: Flinter F, ed. The Genetics of Renal Disease. Oxford, England: Oxford University Press, 2003: 117–146. Amor D, Delatycki MB, Susman M, et al. 46,XX/46,XY at amniocentesis in a fetus with true hermaphroditism. J Med Genet 36: 866–869, 1999.
Amor DJ, Halliday J. A review of known imprinting syndromes and their association with assisted reproduction technologies. Hum Reprod 23: 2826–2834, 2008. Amor DJ, Neo WT, Waters E, Heussler H, Pertile M, Halliday J. Health and developmental outcome of children following prenatal diagnosis of confined placental mosaicism. Prenat Diagn 26: 443–448, 2006. Amor DJ, Woods CG. Pseudotrisomy 13 syndrome in siblings. Clin Dysmorphol 9: 115–118, 2000. Amor DJ, Xu JX , Halliday JL, et al. Pregnancies conceived using assisted reproductive technologies (ART) have low levels of pregnancy-associated plasma protein-A (PAPP-A) leading to a high rate of false-positive results in first trimester screening for Down syndrome. Hum Reprod 24: 1330–1338, 2009. Anahory T, Hamamah S, Andreo B, et al. Sperm segregation analysis of a (13;22) Robertsonian translocation carrier by FISH: a comparison of locus-specific probe and whole chromosome painting. Hum Reprod 20: 1850–1854, 2005. Anckaert E, Schiettecatte J, Sleurs E, Devroey P, Smitz J. First trimester screening for Down’s syndrome after assisted reproductive technology: non-male factor infertility is associated with elevated free beta-human chorionic gonadotropin levels at 10–14 weeks of gestation. Fertil Steril 90: 1206–1210, 2008. Anderlid BM, Sahlén S, Schoumans J, et al. Detailed characterization of 12 supernumerary ring chromosomes using micro-FISH and search for uniparental disomy. Am J Med Genet 99: 223–233, 2001. Anderlid BM, Schoumans J, Annerén G, et al. Subtelomeric rearrangements detected in patients with idiopathic mental retardation. Am J Med Genet 107: 275–284, 2002. Anderson G. Nondirectiveness in prenatal genetics: patients read between the lines. Nurs Ethics 6: 126–136, 1999. Ang DC, Rodríguez Urrego PA , Prasad V. Placental mesenchymal dysplasia: a potential misdiagnosed entity. Arch Gynecol Obstet 279: 937–939, 2009. Angell R . First-meiotic-division nondisjunction in human oocytes. Am J Hum Genet 61: 23–32, 1997. Angell RR , Jacobs PA . Lateral asymmetry in human constitutive heterochromatin: frequency and inheritance. Am J Hum Genet 30: 144–152, 1978. Anguiano A , Yang X , Felix JK , Hoo JJ. Twin brothers with MIDAS syndrome and XX karyotype. Am J Med Genet 119A : 47–49, 2003.
REFERENCES
•
513
Annable K , Donnenfeld AE, Fischer RL, Knops J. Prenatal diagnosis of a jumping translocation. Prenat Diagn 28: 767–769, 2008. Annunziato AT. DNA packaging: nucleosomes and cromatin. Nature Education 1, 2008. Antoccia A , Kobayashi J, Tauchi H, Matsuura S, Komatsu K . Nijmegen breakage syndrome and functions of the responsible protein, NBS1. Genome Dyn 1: 191–205, 2006. Anton E, Blanco J, Egozcue J, Vidal F. Risk assessment and segregation analysis in a pericentric inversion inv6p23q25 carrier using FISH on decondensed sperm nuclei. Cytogenet Genome Res 97: 149–154, 2002. Anton E, Blanco J, Egozcue J, Vidal F. Sperm studies in heterozygote inversion carriers: a review. Cytogenet Genome Res 111: 297–304, 2005. Antonarakis SE, Avramopoulos D, Blouin JL, Talbot CC, Schinzel AA . Mitotic errors in somatic cells cause trisomy 21 in about 4.5% of cases and are not associated with advanced maternal age. Nat Genet 3: 146–150, 1993. Antonell A , Del Campo M, Magano LF, et al. Partial 7q11.23 deletions further implicate GTF2I and GTF2IRD1 as the main genes responsible for the Williams-Beuren syndrome neurocognitive profile. J Med Genet 47: 312–320, 2010. Apacik C, Cohen M, Jakobeit M, et al. Two brothers with multiple congenital anomalies and mental retardation due to disomy (X)(q12→q13.3) inherited from the mother. Clin Genet 50: 63–73, 1996. Archer HL, Gupta S, Enoch S, et al. Distinct phenotype associated with a cryptic subtelomeric deletion of 19p13.3-pter. Am J Med Genet 136: 38–44, 2005. Ardalan A , Prieur M, Choiset A , Turleau C, Goutieres F, Girard-Orgeolet S. Intrachromosomal insertion mimicking a pericentric inversion: molecular cytogenetic characterization of a three break rearrangement of chromosome 20. Am J Med Genet 138A : 288–293, 2005. Arens YHJM, Engelen JJM, Govaerts LCP, et al. Familial insertion (3;5)(q25.3;q22.1q31.3) with deletion or duplication of chromosome region 5q22.1–5q31.3 in ten unbalanced carriers. Am J Med Genet 130A : 128–133, 2004. Arlt MF, Casper AM, Glover TW. Common fragile sites. Cytogenet Genome Res 100: 92–100, 2003. Armstrong L, McGowan-Jordan J, Brierley K , Allanson JE. De novo dup(X)(q22.3q26) in a girl with evidence that functional disomy of X material is the cause of her abnormal phenotype. Am J Med Genet 116A : 71–76, 2003.
514
•
REFERENCES
Arnon J, Meirow D, Lewis-Roness H, Ornoy A . Genetic and teratogenic effects of cancer treatments on gametes and embryos. Hum Reprod Update 7: 394–403, 2001. Asada H, Sueoka K , Hashiba T, Kuroshima M, Kobayashi N, Yoshimura Y. The effects of age and abnormal sperm count on the nondisjunction of spermatozoa. J Assist Reprod Genet 17: 51–59, 2000. Ashley T. Prediction of mammalian meiotic synaptic and recombinational behavior of inversion heterozygotes based on mitotic breakpoint data and the possible evolutionary consequences. Genetica 83: 1–7, 1990. Ashley T, Gaeth AP, Inagaki H, et al. Meiotic recombination and spatial proximity in the etiology of the recurrent t(11;22). Am J Hum Genet 79: 524–538, 2006. Ashton-Prolla P, Gershin IF, Babu A , et al. Prenatal diagnosis of a familial interchromosomal insertion of Y chromosome heterochromatin. Am J Med Genet 73: 470–473, 1997. Atkins KE, Gregg A , Spikes AS, et al. Identification of Y chromatin directly in gonadal tissue by fluorescence in situ hybridization (FISH): significance for Ullrich-Turner syndrome screening in the cytogenetics laboratory. Am J Med Genet 91: 377–382, 2000. Auerbach AD, Min Z, Ghosh R , et al. Clastogeninduced chromosomal breakage as a marker for first trimester prenatal diagnosis of Fanconi anemia. Hum Genet 73: 86–88, 1986. Aughton DJ, AlSaadi AA , Canady AI, Lucas BM. Balanced reciprocal translocation mosaicism associated with an abnormal phenotype. Am J Med Genet 45: 721–724, 1993. Austad T. The right not to know—worthy of preservation any longer? An ethical perspective. Clin Genet 50: 85–88, 1996. Avenarius MR , Hildebrand MS, Zhang Y, et al. Human male infertility caused by mutations in the CATSPER1 channel protein. Am J Hum Genet 84: 505–510, 2009. Aviram-Goldring A , Daniely M, Dorf H, Chaki R , Goldman B, Barkai G. Use of interphase fluorescence in situ hybridization in third trimester fetuses with anomalies and growth retardation. Am J Med Genet 87: 203–206, 1999. Aviv A , Aviv H. Telomeres, hidden mosaicism, loss of heterozygosity, and complex genetic traits. Hum Genet 103: 2–4, 1998. Awa AA , Honda T, Neriishi S, et al. Cytogenetic study of the offspring of atomic bomb survivors, Hiroshima and Nagasaki. In: Obe G, Basler A. eds.
Cytogenetics: Basic and Applied Aspects. Berlin: Springer-Verlag , 1987. Axt-Fliedner R , Hartge D, Chiriac A , et al. Long-term outcome for children born after a first-trimester measurement of increased nuchal translucency with a normal karyotype: a retrospective analysis. Ultraschall Med 30: 558–563, 2009. Aydemir N, Bilaloglu R . The cytogenetic effects of pyrimethamine on male mouse germ cells. J Environ Pathol Toxicol Oncol 15: 79–83, 1996. Aytoz A , De Catte L, Camus M, et al. Obstetric outcome after prenatal diagnosis in pregnancies obtained after intracytoplasmic sperm injection. Hum Reprod 13: 2958–2961, 1998. Ayukawa H, Tsukahara M, Fukuda M, Kondoh O. Recombinant chromosome 18 resulting from a maternal pericentric inversion. Am J Med Genet 50: 323–325, 1994. Baarends WM, van der Laan R , Grootegoed JA . DNA repair mechanisms and gametogenesis. Reproduction 121: 31–39, 2001. Baart EB, Martini E, Eijkemans MJ, et al. Milder ovarian stimulation for in-vitro fertilization reduces aneuploidy in the human preimplantation embryo: a randomized controlled trial. Hum Reprod 22: 980–988, 2007. Baart EB, Martini E, Van Opstal D. Screening for aneuploidies of ten different chromosomes in two rounds of FISH: a short and reliable protocol. Prenat Diagn 24: 955–961, 2004. Babovic-Vuksanovic D, Jenkins SC, Ensenauer R , Newman DC, Jalal SM. Subtelomeric deletion of 18p in an adult with paranoid schizophrenia and mental retardation. Am J Med Genet 124A : 318–322, 2004. Babu A , Agarwal AK , Verma RS. A new approach in recognition of heterochromatic regions of human chromosomes by means of restriction endonucleases. Am J Hum Genet 42: 60–65, 1988. Baccetti B, Bruni E, Collodel G, et al. 10, 15 reciprocal translocation in an infertile man: ultrastructural and fluorescence in-situ hybridization sperm study: case report. Hum Reprod 18: 2302–2308, 2003. Baccetti B, Capitani S, Collodel G, Estenoz M, Gambera L, Piomboni P. Infertile spermatozoa in a human carrier of robertsonian translocation 14;22. Fertil Steril 78: 1127–1130, 2002. Baccetti B, Collodel G, Marzella R , et al. Ultrastructural studies of spermatozoa from infertile males with Robertsonian translocations and 18, X, Y aneuploidies. Hum Reprod 20: 2295–2300, 2005. Bache I, Brondum-Nielsen K , Tommerup N. Genetic counseling in adult carriers of a balanced
chromosomal rearrangement ascertained in childhood: experiences from a nationwide reexamination of translocation carriers. Genet Med 9: 185–187, 2007. Bache I, Hjorth M, Bugge M, et al. Systematic re-examination of carriers of balanced reciprocal translocations: a strategy to search for candidate regions for common and complex diseases. Eur J Hum Genet 14: 410–417, 2006. Bachmann-Gagescu R , Mefford HC, Cowan C, et al. Recurrent 200-kb deletions of 16p11.2 that include the SH2B1 gene are associated with developmental delay and obesity. Genet Med 12: 641–647, 2010. Baena N, De Vigan C, Cariati E, et al. Prenatal detection of rare chromosomal autosomal abnormalities in Europe. Am J Med Genet 118A : 319–327, 2003. Baena N, De Vigan C, Cariati E, et al. Turner syndrome: evaluation of prenatal diagnosis in 19 European registries. Am J Med Genet 129A : 16–20, 2004. Bagshawe KD, Lawler SD. Unmasking moles. Br J Obstet Gynaecol 89: 255–257, 1982. Bahçe M, Cohen J, Munné S. Preimplantation genetic diagnosis of aneuploidy: were we looking at the wrong chromosomes? J Assist Reprod Genet 16: 176–181, 1999. Bahçe M, Oõur G, Ýmirzalýoõlu N, et al. Repeated fetal loss in two related first cousin marriages with couples all carrying the same translocation t(13q;14q). Eur J Hum Genet 4: 35–36, 1996. Baillie M. (1799) A series of engravings, accompanied with explanations, which are intended to illustrate the morbid anatomy of some of the most important parts of the human body. Ninth fasciculus. W. Bulmer and Co, London. Facsimile edition prep. H Attwood, Melbourne University Press, 1985. Baker E, Hinton L, Callen DF, et al. Study of 250 children with idiopathic mental retardation reveals nine cryptic and diverse subtelomeric chromosome anomalies. Am J Med Genet 107: 285–293, 2002. Baker PR , Tsai AC, Springer M, et al. Male with mosaicism for supernumerary ring X chromosome: analysis of phenotype and characterization of genotype using array comparative genome hybridization. J Craniofac Surg 21:1369–1375, 2010. Bakhshi S, Joenje H, Schindler D, et al. A case report of a patient with microcephaly, facial dysmorphism, mitomycin-c-sensitive lymphocytes, and susceptibility to lymphoma. Cancer Genet Cytogenet 164: 168–171, 2006.
REFERENCES
•
515
Bakshi SR , Dave BJ, Sanger W, et al. Characterization of a familial small supernumerary marker chromosome in a patient with adult-onset tongue cancer. Cytogenet Genome Res 121: 14–17, 2008. Balakier H, Bouman D, Sojecki A , Librach C, Squire JA . Morphological and cytogenetic analysis of human giant oocytes and giant embryos. Hum Reprod 17: 2394–2401, 2002. Balasubramanian M, Smith K , Basel-Vanagaite L, et al. Case series: 2q33.1 microdeletion syndrome— further delineation of the phenotype. J Med Genet 48: 290–298, 2011. Balcı S, Engiz Ö, Aktaş D, et al. Ring chromosome 4 and Wolf-Hirschhorn syndrome (WHS) in a child with multiple anomalies. Am J Med Genet 140: 628–632, 2006. Balci S, Tümer C, Karaca C, Bartsch O. Familial ring (18) mosaicism in a 23-year-old young adult with 46,XY,r(18) (::p11→q21::)/46,XY karyotype, intellectual disability, motor retardation and single maxillary incisor and in his phenotypically normal mother, karyotype 47,XX,+r(18) (::p11→q21::)/46,XX . Am J Med Genet 155A : 1129–1135, 2011. Balcı S, Unal A , Engiz O, et al. Bilateral periventricular nodular heterotopia, severe learning disability, and epilepsy in a male patient with 46,XY,der(19) t(X;19) (q11.1–11.2;p13.3). Dev Med Child Neurol 49: 219–224, 2007. Baldwin EL, May LF, Justice AN, Martin CL, Ledbetter DH. Mechanisms and consequences of small supernumerary marker chromosomes: from Barbara McClintock to modern genetic-counseling issues. Am J Hum Genet 82: 398–410, 2008. Balikova I, Martens K , Melotte C, et al. Autosomaldominant microtia linked to five tandem copies of a copy-number-variable region at chromosome 4p16. Am J Hum Genet 82: 181–187, 2008. Balikova I, Menten B, de Ravel T, et al. Subtelomeric imbalances in phenotypically normal individuals. Hum Mutat 28: 958–967, 2007. Ballarati L, Recalcati MP, Bedeschi MF, et al. Cytogenetic, FISH and array-CGH characterization of a complex chromosomal rearrangement carried by a mentally and language impaired patient. Eur J Med Genet 52: 218–223, 2009. Ballif BC, Hornor SA, Jenkins E, et al. Discovery of a previously unrecognized microdeletion syndrome of 16p11.2-p12.2. Nat Genet 39: 1071–1073, 2007a. Ballif BC, Hornor SA , Sulpizio SG, et al. Development of a high-density pericentromeric region BAC clone set for the detection and characterization of small supernumerary marker chromosomes by array CGH. Genet Med 9: 150–162, 2007b.
516
•
REFERENCES
Ballif BC, Kashork CD, Shaffer LG. The promise and pitfalls of telomere region-specific probes. Am J Hum Genet 67: 1356–1359, 2000a. Ballif BC, Kashork CD, Shaffer LG. FISHing for mechanisms of cytogenetically defined terminal deletions using chromosome-specific subtelomeric probes. Eur J Hum Genet 8: 764–770, 2000b. Ballif BC, Rorem EA , Sundin K , et al. Detection of low-level mosaicism by array CGH in routine diagnostic specimens. Am J Med Genet 140: 2757–2767, 2006. Ballif BC, Sulpizio SG, Lloyd RM, et al. The clinical utility of enhanced subtelomeric coverage in array CGH. Am J Med Genet 143A : 1850–1857, 2007c. Ballif BC, Theisen A , Coppinger J, et al. Expanding the clinical phenotype of the 3q29 microdeletion syndrome and characterization of the reciprocal microduplication. Mol Cytogenet 1: 8, 2008. Ballif BC, Theisen A , Rosenfeld JA, et al. Identification of a recurrent microdeletion at 17q23.1q23.2 flanked by segmental duplications associated with heart defects and limb abnormalities. Am J Hum Genet 86: 454–461, 2010. Ballif BC, Yu W, Shaw CA , Kashork CD, Shaffer LG. Monosomy 1p36 breakpoint junctions suggest pre-meiotic breakage-fusion-bridge cycles are involved in generating terminal deletions. Hum Mol Genet 12: 2153–2165, 2003. Balmer D, Baumer A , Röthlisberger B, Schinzel A . Severe intra-uterine growth retardation in a patient with maternal uniparental disomy 22 and a 22-trisomic placenta. Prenat Diagn 19: 1061–1064, 1999. Bandyopadhyay R , Berend SA , Page SL, Choo KH, Shaffer LG. Satellite III sequences on 14p and their relevance to Robertsonian translocation formation. Chromosome Res 9: 235–242, 2001a. Bandyopadhyay R , Heller A , Knox-DuBois C, et al. Parental origin and timing of de novo Robertsonian translocation formation. Am J Hum Genet 71: 1456–1462, 2002. Bandyopadhyay R , McCaskill C, Knox-Du Bois C, et al. Mosaicism in a patient with Down syndrome reveals post-fertilization formation of a Robertsonian translocation and isochromosome. Am J Med Genet 116A : 159–163, 2003. Bandyopadhyay R , McQuillan C, Page SL, Choo KHA , Shaffer LG. Identification and characterization of satellite III subfamilies to the acrocentric chromosomes. Chromosome Res 9: 223–233, 2001b. Banerjee I, Shevlin M, Taranissi M, et al. Health of children conceived after preimplantation genetic diagnosis: a preliminary outcome study. Reprod Biomed Online 16: 376–381, 2008.
Baptista J, Mercer C, Prigmore E, et al. Breakpoint mapping and array CGH in translocations: comparison of a phenotypically normal and an abnormal cohort. Am J Hum Genet 82: 927–936, 2008. Baptista MJ, Fairbrother UL, Howard CM, et al. Heterotrisomy, a significant contributing factor to ventricular septal defect associated with Down syndrome? Hum Genet 107: 476–482, 2000. Baralle D, Willatt LR , Shears DJ. Leri-Weill syndrome associated with a pseudodicentric X;Y translocation chromosome and skewed X-inactivation: implications for genetic counselling. Am J Med Genet 95: 391–395, 2000. Barbaro M, Cicognani A , Balsamo A , et al. Gene dosage imbalances in patients with 46,XY gonadal DSD detected by an in-house-designed synthetic probe set for multiplex ligation-dependent probe amplification analysis. Clin Genet 73: 453–464, 2008. Barbash-Hazan S, Frumkin T, Malcov M, et al. Preimplantation aneuploid embryos undergo self-correction in correlation with their developmental potential. Fertil Steril 92: 890–896, 2009. Barber JCK . Directly transmitted unbalanced chromosome abnormalities and euchromatic variants. J Med Genet 42: 609–629, 2005. Barber JCK . Chromosomal copy number variation. Europ Cytogenet Assoc Newsletter 22: 9, 2008a. Barber JCK . Terminal 3p deletions: phenotypic variability, chromosomal non-penetrance, or gene modification? Am J Med Genet 146A : 1899–1901, 2008b Barber JCK . Why a causal association between classical euchromatic variants of chromosome 9 and multiple miscarriages is unlikely. Genet Couns 21: 469–471, 2010. Barber JCK , Bunyan D, Curtis M, et al. 8p23.1 duplication syndrome differentiated from copy number variation of the defensin cluster at prenatal diagnosis in four new families. Mol Cytogenet 3: 3, 2010a. Barber JCK , Cockwell AE, Grant E, Williams S, Dunn R , Ogilvie CM. Is karyotyping couples experiencing recurrent miscarriage worth the cost? Br J Obstet Gynaecol 117: 885–888, 2010b. Barber JCK , Ellis KH, Bowles LV, et al. Adenomatous polyposis coli and a cytogenetic deletion of chromosome 5 resulting from a maternal intrachromosomal insertion. J Med Genet 31: 312–316, 1994. Barber JCK , Joyce CA , Collinson MN, et al. Duplication of 8p23. 1: a cytogenetic anomaly
with no established clinical significance. J Med Genet 35: 491–496, 1998. Barber JCK , Maloney V, Hollox EJ, et al. Duplications and copy number variants of 8p23.1 are cytogenetically indistinguishable but distinct at the molecular level. Eur J Hum Genet 13: 1131–1136, 2005a. Barber JCK , Maloney VK , Huang S, et al. 8p23.1 duplication syndrome; a novel genomic condition with unexpected complexity revealed by array CGH. Eur J Hum Genet 16: 18–27, 2008. Barber JCK , Maloney VK , Kirchhoff M, Thomas NS, Boyle TA , Castle B. Transmitted duplication of 12q21.32–12q22 includes 48 genes and has no apparent phenotypic consequences. Am J Med Genet 143: 615–618, 2007. Barber JCK , Thomas NS, Collinson MN, et al. Segmental haplosufficiency: transmitted deletions of 2p12 include a pancreatic regeneration gene cluster and have no apparent phenotypic consequences. Eur J Hum Genet 13: 283–291, 2005b. Barber JCK , Zhang S, Friend N, et al. Duplications of proximal 16q flanked by heterochromatin are not euchromatic variants and show no evidence of heterochromatic position effect. Cytogenet Genome Res 114: 351–358, 2006. Barbosa AS, Ferraz-Costa TE, Semer M, Liberman B, Moreira-Filho CA . XY gonadal dysgenesis and gonadoblastoma: a study in two sisters with a cryptic deletion of the Y chromosome involving the SRY gene. Hum Genet 95: 63–66, 1995. Bardhan A. Many functions of the meiotic cohesin. Chromosome Res 18: 909–24, 2010. Barge-Schaapveld DQCM, Maas SM, Polstra A , Knegt LC, Hennekam RCM. The atypical 16p11.2 deletion: A not so atypical microdeletion syndrome? Am J Med Genet 155A : 1066–1072, 2011. Baris HN, Tan WH, Kimonis VE, Irons MB. Diagnostic utility of array-based comparative genomic hybridization in a clinical setting. Am J Med Genet 143A :2523–2533, 2007. Barišić I, Zergollern L, Mužinić D, Hitrec V. Risk estimates for balanced reciprocal translocation carriers—prenatal diagnosis experience. Clin Genet 49: 145–151, 1996. Barlow AL, Hultén MA . Combined immunocytogenetic and molecular cytogenetic analysis of meiosis I human spermatocytes. Chromosome Res 4: 562–573, 1996. Barlow GM, Micales B, Lyons GE, Korenberg JR . Down syndrome cell adhesion molecule is conserved in mouse and highly expressed in the
REFERENCES
•
517
adult mouse brain. Cytogenet Cell Genet 94: 155–162, 2001. Barseghyan K , Sklansky MS, Paquette LB, Randolph LM, Miller DA . Agenesis of the ductus venosus in a fetus with nonmosaic trisomy 22. Prenat Diagn 29: 901–902, 2009. Bartels I, Schlueter G, Liehr T, et al. Supernumerary small marker chromosome (SMC) and uniparental disomy 22 in a child with confined placental mosaicism of trisomy 22: trisomy rescue due to marker chromosome formation. Cytogenet Genome Res 101: 103–105, 2003. Bartels I, Starke H, Argyriou L, Sauter SM, Zoll B, Liehr T. An exceptional complex chromosomal rearrangement (CCR) with eight breakpoints involving four chromosomes (1;3;9;14) in an azoospermic male with normal phenotype. Eur J Med Genet 50: 133–138, 2007. Bartholdi D, Krajewska-Walasek M, Őunap K , et al. Epigenetic mutations of the imprinted IGF2-H19 domain in Silver-Russell syndrome (SRS): results from a large cohort of patients with SRS and SRS-like phenotypes. J Med Genet 46: 192–197, 2009. Bartsch O, Gebauer K , Lechno S, et al. Four unrelated patients with Lubs X-linked mental retardation syndrome and different Xq28 duplications. Am J Med Genet 152A : 305–312, 2010. Bartsch O, König U, Petersen MB, et al. Cytogenetic, FISH and DNA studies in 11 individuals from a family with two siblings with dup(21q) Down syndrome. Hum Genet 92: 127–132, 1993. Bartsch O, Nemecková M, Kocárek E, et al. DiGeorge/velocardiofacial syndrome: FISH studies of chromosomes 22q11 and 10p14, and clinical reports on the proximal 22q11 deletion. Am J Med Genet 117A : 1–5, 2003. Bartsch O, Wuyts W, Van Hul W, et al. Delineation of a contiguous gene syndrome with multiple exostoses, enlarged parietal foramina, craniofacial dysostosis, and mental retardation, caused by deletions in the short arm of chromosome 11. Am J Hum Genet 58: 734–742, 1996. Basehore MJ, Friez MJ. Molecular analysis of Fragile X syndrome. Curr Protoc Hum Genet 9: 5, 2009. Basel D, Kilpatrick MW, Tsipouras P. The expanding panorama of split hand foot malformation. Am J Med Genet 140: 1359–1365, 2006. Basel-Vanagaite L, Davidov B, Friedman J, et al. Amniotic trisomy 11 mosaicism—is it a benign finding? Prenat Diagn 26: 778–781, 2006. Basinko A , Douet-Guilbert N, Parent P, et al. Familial interstitial deletion of the short arm of chromosome 4 (p15.33-p16.3) characterized by
518
•
REFERENCES
molecular cytogenetic analysis. Am J Med Genet 146: 899–903, 2008. Bass HN, Sparkes RS, Lessner MM, Fox M, Phoenix B, Bernar J. A family with three independent autosomal translocations associated with 7q32→7qter syndrome. J Med Genet 22: 59–63, 1985. Bastepe M. The GNAS locus and pseudohypoparathyroidism. Adv Exp Med Biol 626: 27–40, 2008. Bates AW. Autopsy on a case of Roberts syndrome reported in 1672: the earliest description? Am J Med Genet 117A : 92–96, 2003. Batista DAS, Pai GS, Stetten G. Molecular analysis of a complex chromosomal rearrangement and a review of familial cases. Am J Med Genet 53: 255–263, 1994. Batstone PJ, Simpson S, Bonthron DT, et al. Effective monosomy or trisomy of chromosome band 2q37.3 due to the unbalanced segregation of a 2;11 translocation. Am J Med Genet 118A : 241–246, 2003. Battaglia A . The inv dup (15) or idic (15) syndrome (Tetrasomy 15q). Orphanet J Rare Dis 3: 30, 2008. Battaglia A , Carey JC. Health supervision and anticipatory guidance of individuals with Wolf-Hirschhorn syndrome. Am J Med Genet 89: 111–115, 1999. Battaglia A , Hoyme HE, Dallapiccola B, et al. Further delineation of deletion 1p36 syndrome in 60 patients: a recognizable phenotype and common cause of developmental delay and mental retardation. Pediatrics 121: 404–410, 2008. Battaglia DE, Goodwin P, Klein NA , Soules MR . Influence of maternal age on meiotic spindle assembly in oocytes from naturally cycling women. Hum Reprod 11: 2217–2222, 1996. Baty BJ, Blackburn BL, Carey JC. Natural history of trisomy 18 and trisomy 13: I. Growth, physical assessment, medical histories, survival, and recurrence risk. Am J Med Genet 49: 175–188, 1994. Baty BJ, Olson SB, Magenis RE, Carey JC. Trisomy 20 mosaicism in two unrelated girls with skin hypopigmentation and normal intellectual development. Am J Med Genet 99: 210–216, 2001. Baud O, Cormier-Daire V, Lyonnet S, Desjardins L, Turleau C, Doz F. Dysmorphic phenotype and neurological impairment in 22 retinoblastoma patients with constitutional cytogenetic 13q deletion. Clin Genet 55: 478–482, 1999. Baumann W, Zabel B, Holl M. Familial pericentric inversion of the X chromosome [inv(X) (p11q28)]. Ann Génét 27: 106–108, 1984.
Baumer A , Basaran S, Taralczak M, et al. Initial maternal meiotic I error leading to the formation of a maternal i(2q) and a paternal i(2p) in a healthy male. Cytogenet Genome Res 118: 38–41, 2007. Baumer A , Dres D, Basaran S, Isci H, Dehgan T, Schinzel A . Parental origin of the two additional haploid sets of chromosomes in an embryo with tetraploidy. Cytogenet Genome Res 101: 5–7, 2003. Baumgartner A , Schmid TE, Schuetz CG, Adler ID. Detection of aneuploidy in rodent and human sperm by multicolor FISH after chronic exposure to diazepam. Mutat Res 490: 11–19, 2001. Baysal BE, Potkin SG, Farr JE, et al. Bipolar affective disorder partially cosegregates with a balanced t(9;11)(p24;q23.1) chromosomal translocation in a small pedigree. Am J Med Genet 81: 81–91, 1998. Bedoyan JK , Flore LA , Alkatib A , Ebrahim SA , Bawle EV. Transmission of ring chromosome 13 from a mother to daughter with both having a 46,XX, r(13)(p13q34) karyotype. Am J Med Genet 129A : 316–320, 2004. Beekhuis JR , De Wolf BT, Mantingh A , Heringa MP. The influence of serum screening on the amniocentesis rate in women of advanced maternal age. Prenat Diagn 14: 199–202, 1994. Bejjani BA , Shaffer LG. Clinical utility of contemporary molecular cytogenetics. Annu Rev Genomics Hum Genet 9: 71–86, 2008. Bekker MN, van den Akker NM, Bartelings MM, et al. Nuchal edema and venous-lymphatic phenotype disturbance in human fetuses and mouse embryos with aneuploidy. J Soc Gynecol Investig 13: 209–216, 2006. Belin V, Farhat M, Monset-Couchard M. Intracytoplasmic sperm injection pregnancy with trisomy 20p and monosomy 22q in a newborn resulting from a balanced paternal translocation. Biol Neonate 75: 398–401, 1999. Bell J, Dunlop R , Bryan J. Another paracentric inversion of chromosome 18. Am J Med Genet 39: 238, 1991. Bendavid C, Rochard L, Dubourg C, et al. Array-CGH analysis indicates a high prevalence of genomic rearrangements in holoprosencephaly: an updated map of candidate loci. Hum Mutat 30: 1175–1182, 2009. Bender BG, Harmon RJ, Linden MG, BucherBartelson B, Robinson A . Psychosocial competence of unselected young adults with sex chromosome abnormalities. Am J Med Genet 88: 200–206, 1999. Bender BG, Harmon RJ, Linden MG, Robinson A . Psychosocial adaptation of 39 adolescents with sex
chromosome abnormalities. Pediatrics 96: 302–308, 1995. Bender BG, Linden MG, Harmon RJ. Neuropsychological and functional cognitive skills of 35 unselected adults with sex chromosome abnormalities. Am J Med Genet 102: 309–313, 2001. Bender BG, Linden MG, Robinson A . Neuropsychological impairment in 42 adolescents with sex chromosome abnormalities. Am J Med Genet 48: 169–173, 1993. Benet J, Oliver-Bonet M, Cifuentes P, Templado C, Navarro J. Segregation of chromosomes in sperm of reciprocal translocation carriers: a review. Cytogenet Genome Res 111: 281–290, 2005. Benetti E, Murer L, Bordugo A , Andreetta B, Artifoni L. 10p12.1 deletion: HDR phenotype without DGS2 features. Exp Mol Pathol 86: 74–76, 2009. Benkendorf JL, Prince MB, Rose MA , De Fina A , Hamilton HE. Does indirect speech promote nondirective genetic counseling? Results of a sociolinguistic investigation. Am J Med Genet 106: 199–207, 2001. Benkhalifa M, Janny L, Vye P, Malet P, Boucher D, Menezo Y. Assessment of polyploidy in human morulae and blastocysts using co-culture and fluorescent in-situ hybridization. Hum Reprod 8: 895–902, 1993. Benlian P, Foubert L, Gagne E, et al. Complete paternal isodisomy for chromosome 8 unmasked by lipoprotein lipase deficiency. Am J Hum Genet 59: 431–436, 1996. Benn P. Trisomy 16 and trisomy 16 Mosaicism: a review. Am J Med Genet 79: 121–133, 1998. Benn PA , Hsu LYF. Evidence for preferential involvement of chromosome bands 6p21 and 13q14 in amniotic fluid cell balanced translocation pseudomosaicism. Clin Genet 29: 116–121, 1986. Ben-Shachar S, Lanpher B, German JR , et al. Microdeletion 15q13. 3: a locus with incomplete penetrance for autism, mental retardation, and psychiatric disorders. J Med Genet 46: 382–388, 2009. Ben-Shachar S, Ou Z, Shaw CA , et al. 22q11.2 distal deletion: a recurrent genomic disorder distinct from DiGeorge syndrome and velocardiofacial syndrome. Am J Hum Genet 82: 214–221, 2008. Betancur C. Etiological heterogeneity in autism spectrum disorders: More than 100 genetic and genomic disorders and still counting. Brain Res 1380:42–77, 2011. Bentley RW, Pearson J, Gearry RB, et al. Association of higher DEFB4 genomic copy number with Crohn’s disease. Am J Gastroenterol 105: 354–359, 2010.
REFERENCES
•
519
Benzacken B, Gavelle FM, Martin-Pont B, et al. Familial sperm polyploidy induced by genetic spermatogenesis failure: case report. Hum Reprod 16: 2646–2651, 2001a. Benzacken B, Monier-Gavelle F, Siffroi JP, Agbo P, Chalvon A , Wolf JP. Acrocentric chromosome polymorphisms: beware of cryptic translocations. Prenat Diagn 21: 96–98, 2001b. Berend SA , Bejjani BA , McCaskill C, Shaffer LG. Identification of uniparental disomy in phenotypically abnormal carriers of isochromosomes or Robertsonian translocations. Am J Med Genet 111: 362–365, 2002a. Berend SA , Bodamer OAF, Shapira SK , Shaffer LG, Bacino CA . Familial complex chromosomal rearrangement resulting in a recombinant chromosome. Am J Med Genet 109: 311–317, 2002b. Berend SA , Canun S, McCaskill C, Page SL, Shaffer LG. Molecular analysis of mosaicism for two different de novo acrocentric rearrangements demonstrates diversity in Robertsonian translocation formation. Am J Med Genet 80: 252–259, 1998. Berend SA , Feldman GL, McCaskill C, Czarnecki P, Van Dyke DL, Shaffer LG. Investigation of two cases of paternal disomy 13 suggests timing of isochromosome formation and mechanisms leading to uniparental disomy. Am J Med Genet 82: 275–281, 1999. Berend SA , Horwitz J, McCaskill C, Shaffer LG. Identification of uniparental disomy following prenatal detection of Robertsonian translocations and isochromosomes. Am J Hum Genet 66: 1787–1793, 2000a. Berend SA , Spikes AS, Kashork CD, et al. Dual-probe fluorescence in situ hybridization assay for detecting deletions associated with VCFS/ DiGeorge syndrome I and DiGeorge syndrome II loci. Am J Med Genet 91: 313–317, 2000b. Berg FD, Kurzl R , Hinrichsen MJ, Zander J. Familial 46,XY pure gonadal dysgenesis and gonadoblastoma/dysgerminoma: case report. Gynecol Oncol 32: 261–267, 1989. Berg JM, Korossy M. Down syndrome before Down: a retrospect. Am J Med Genet 102: 205–211, 2001. Bergemann E. Manifestation familiale du karyotype triplo-X . Communication préliminaire. J Génét Hum 10: 370–371, 1962. Berger SL, Kouzarides T, Shiekhattar R , Shilatifard A . An operational definition of epigenetics. Genes Dev 23: 781–783, 2009. Bergère M, Wainer R , Nataf V, et al. Biopsied testis cells of four 47,XXY patients: fluorescence in-situ
520
•
REFERENCES
hybridization and ICSI results. Hum Reprod 17: 32–37, 2002. Berghella V, Wapner RJ, Yang-Feng T, Mahoney MJ. Prenatal confirmation of true fetal trisomy 22 mosaicism by fetal skin biopsy following normal fetal blood sampling. Prenat Diagn 18: 384–389, 1998. Bergman A , Blennow E. Inv dup(22), del(22)(q11) and r(22) in the father of a child with DiGeorge syndrome. Eur J Hum Genet 8: 801–804, 2000. Béri-Deixheimer M, Gregoire MJ, Toutain A , et al. Genotype-phenotype correlations to aid in the prognosis of individuals with uncommon 20q13.33 subtelomere deletions: a collaborative study on behalf of the “association des Cytogénéticiens de langue Française.” Eur J Hum Genet 15: 446–452, 2007. Berkenstadt M, Ries-Levavi L, Cuckle H, Peleg L, Barkai G. Preconceptional and prenatal screening for fragile X syndrome: experience with 40,000 tests. Prenat Diagn 27: 991–994, 2007. Berkowitz RS, Bernstein MR , Laborde O, Goldstein DP. Subsequent pregnancy experience in patients with gestational trophoblastic disease. New England Trophoblastic Disease Center, 1965–1992. J Reprod Med 39: 228–232, 1994. Bernardini L, Capalbo A , D’Avanzo MG, et al. Five cases of supernumerary small ring chromosomes 1: heterogeneity and genotype-phenotype correlation. Eur J Med Genet 50: 94–102, 2007. Bernasconi F, Karagüzel A , Celep F, et al. Normal phenotype with maternal isodisomy in a female with two isochromosomes: i(2p) and i(2q). Am J Hum Genet 59: 1114–1118, 1996. Bernert J, Bartels I, Gatz G, et al. Prenatal diagnosis of the Pallister-Killian mosaic aneuploidy syndrome by CVS. Am J Med Genet 42: 747–750, 1992. Berr C, Borghi E, Rethoré MO, Lejeune J, Alperovitch A . Risk of Down syndrome in relatives of trisomy 21 children. A case-control study. Ann Génét 33: 137–140, 1990. Berry-Kravis E, Hessl D, Coffey S, et al. A pilot open label, single dose trial of fenobam in adults with fragile X syndrome. J Med Genet 46: 266–271, 2009. Bersu ET. Anatomical analysis of the developmental effects of aneuploidy in man: the Down syndrome. Am J Med Genet 5: 399–420, 1980. Bertini V, Valetto A , Uccelli A , et al. Molecular cytogenetic characterization of a de novo mosaic supernumerary ring chromosome 7: report of a new patient. Am J Med Genet 146A : 2955–2959, 2008a. Berwick M, Satagopan JM, Ben-Porat L, et al. Genetic heterogeneity among Fanconi anemia
heterozygotes and risk of cancer. Cancer Res 67: 9591–9596, 2007. Bettio D, Baldwin EL, Carrozzo R , et al. Molecular cytogenetic and clinical findings in a patient with a small supernumerary r(8) mosaicism. Am J Med Genet 146A : 247–250, 2008. Bettio D, Levi Setti P, Bianchi P, Grazioli V. Trisomy 18 mosaicism in a woman with normal intelligence. Am J Med Genet 120A : 303–304, 2003. Betts DR , Greiner J, Feldges A , Caflisch U, Niggli FK . Constitutional balanced chromosomal rearrangements and neoplasm in children. J Pediatr Hematol Oncol 23: 582–584, 2001. Betz A , Turleau C, De Grouchy J. Heterozygosity and homozygosity for a pericentric inversion of human chromosone 3. Ann Génét 17: 79–80, 1974. Beunders G, van de Kamp JM, Veenhoven RH, van Hagen JM, Nieuwint AW, Sistermans EA . A triplication of the Williams-Beuren syndrome region in a patient with mental retardation, a severe expressive language delay, behavioural problems and dysmorphisms. J Med Genet 47: 271–275, 2010. Beverstock GC, Hansson K , Helderman-van den Enden AT, et al. A near false-negative finding of mosaic trisomy 21—a cautionary tale. Prenat Diagn 18: 742–746, 1998. Bianca S, Boemi G, Barrano B, et al. Mosaic trisomy 20: considerations for genetic counseling. Am J Med Genet 146A : 1897–1898, 2008. Bianchi DW. Fetomaternal cell trafficking: a new cause of disease? Am J Med Genet 91: 22–28, 2000. Bianchi DW, Hanson J. Sharpening the tools: a summary of a National Institutes of Health workshop on new technologies for detection of fetal cells in maternal blood for early prenatal diagnosis. J Matern Fetal Neonatal Med 19: 199–207, 2006. Bianchi DW, Wilkins-Haug LE, Enders AC, Hay ED. Origin of extraembryonic mesoderm in experimental animals: relevance to chorionic mosaicism in humans. Am J Med Genet 46: 542–550, 1993. Bianco K , Caughey AB, Shaffer BL, Davis R , Norton ME. History of miscarriage and increased incidence of fetal aneuploidy in subsequent pregnancy. Obstet Gynecol 107: 1098–1102, 2006. Biason-Lauber A , Konrad D, Meyer M, DeBeaufort C, Schoenle EJ. Ovaries and female phenotype in a girl with 46,XY karyotype and mutations in the CBX2 gene. Am J Hum Genet 84: 658–663, 2009. Bibi G, Malcov M, Yuval Y, et al. The effect of CGG repeat number on ovarian response among fragile X premutation carriers undergoing
preimplantation genetic diagnosis. Fertil Steril 94: 869–874, 2010. Bieber FR , Nance WE, Morton CC, et al. Genetic studies of an acardiac monster: evidence of polar body twinning in man. Science 213: 775–777, 1981. Bielanska M, Tan SL, Ao A . High rate of mixoploidy among human blastocysts cultured in vitro. Fertil Steril 78: 1248–1253, 2002a. Bielanska M, Tan SL, Ao A . Chromosomal mosaicism throughout human preimplantation development in vitro: incidence, type, and relevance to embryo outcome. Hum Reprod 17: 413–419, 2002b. Biesecker LG, Rosenberg M, Dziadzio L, et al. Detection of a subtle rearrangement of chromosome 22 using molecular techniques. Am J Med Genet 58: 389–394, 1995. Bijlsma EK , Gijsbers AC, Schuurs-Hoeijmakers JH, et al. Extending the phenotype of recurrent rearrangements of 16p11. 2: deletions in mentally retarded patients without autism and in normal individuals. Eur J Med Genet 52: 77–87, 2009. Bilimoria KY, Rothenberg JM. Prenatal diagnosis of a trisomy 7/maternal uniparental heterodisomy 7 mosaic fetus. Am J Med Genet 118A : 60–63, 2003. Billuart P, Bienvenu T, Ronce N, et al. Oligophrenin-1 encodes a rhoGAP protein involved in X-linked mental retardation. Nature 392: 923–926, 1998. Bilousova TV, Dansie L, Ngo M, et al. Minocycline promotes dendritic spine maturation and improves behavioural performance in the fragile X mouse model. J Med Genet 46: 94–102, 2009. Binder G, Seidel AK , Martin DD, et al. The endocrine phenotype in silver-russell syndrome is defined by the underlying epigenetic alteration. J Clin Endocrinol Metab 93: 1402–1407, 2008. Bingham PM, Zimmerman RA , McDonald-McGinn D, Driscoll D, Emanuel BS, Zackai E. Enlarged Sylvian fissures in infants with interstitial deletion of chromosome 22q11. Am J Med Genet 74: 538–543, 1997. Bint SM, Mackie Ogilvie C, Flinter FA , Khalaf Y, Scriven PN. Meiotic segregation of Robertsonian translocations ascertained in cleavage-stage embryos—implications for preimplantation genetic diagnosis. Hum Reprod 26: 1575–1584, 2011. Bird TD. Genetic aspects of Alzheimer disease. Genet Med 10: 231–239, 2008. Birkebaek NH, Cruger D, Hansen J, Nielsen J, Bruun-Petersen G. Fertility and pregnancy outcome in Danish women with Turner syndrome. Clin Genet 61: 35–39, 2002.
REFERENCES
•
521
Bischoff FZ, Simpson JL. Endocervical fetal trophoblast for prenatal genetic diagnosis. Curr Opin Obstet Gynecol 18: 216–220, 2006. Bisgaard AM, Kirchhoff M, Nielsen JE, et al. Chromosomal deletion unmasking a recessive disease: 22q13 deletion syndrome and metachromatic leukodystrophy. Clin Genet 75: 175–179, 2009. Bittel DC, Kibiryeva N, Butler MG. Expression of 4 genes between chromosome 15 breakpoints 1 and 2 and behavioral outcomes in Prader–Willi syndrome. Pediatrics 118: e1276–e1283, 2006. Bittel DC, Theodoro MF, Kibiryeva N, Fischer W, Talebizadeh Z, Butler MG. Comparison of X-chromosome inactivation patterns in multiple tissues from human females. J Med Genet 45: 309–313, 2008. Bittencourt MC, Morris MA , Chabod J, et al. Fortuitous detection of uniparental isodisomy of chromosome 6. J Med Genet 34: 77–78, 1997. Björck EJ, Anderlid BM, Blennow E. Maternal isodisomy of chromosome 9 with no impact on the phenotype in a woman with two isochromosomes: i(9p) and i(9q). Am J Med Genet 87: 49–52, 1999. Blackhall FH, Atkinson AD, Maaya MB, et al. Semen cryopreservation, utilisation and reproductive outcome in men treated for Hodgkin’s disease. Br J Cancer 87: 381–384, 2002. Blaicher W, Repa C, Schaller A . Acardiac twin pregnancy: associated with trisomy 2: case report. Hum Reprod 15: 474–475, 2000. Blair J, Tolmie J, Hollman AS, Donaldson MD. Phenotype, ovarian function, and growth in patients with 45,X/47,XXX Turner mosaicism: implications for prenatal counseling and estrogen therapy at puberty. J Pediatr 139: 724–728, 2001. Blancato JK . Fluorescence in situ hybridization. In: Gersen SL, Keagle MB, eds. The Principles of Clinical Cytogenetics. Totowa, NJ: Humana Press, 1999: 443–471. Blanco J, Gabau E, Gomez D, et al. Chromosome 21 disomy in the spermatozoa of the fathers of children with trisomy 21, in a population with a high prevalence of Down syndrome: increased incidence in cases of paternal origin. Am J Hum Genet 63: 1067–1072, 1998. Blennow E, Nielsen KB, Telenius H, et al. Fifty probands with extra structurally abnormal chromosomes characterized by fluorescence in situ hybridization. Am J Med Genet 55: 85–94, 1995. Blennow E, Sahlén S, Inzunza J, Hovatta O, Nordenskjöld M, Malmgren H. Single cell CGH analysis of human preimplantation embryos from
522
•
REFERENCES
PGD patients with balanced structural chromosome aberrations. Ann Génét 44: s26, 2001. Bliek J, Snijder S, Maas SM, et al. Phenotypic discordance upon paternal or maternal transmission of duplications of the 11p15 imprinted regions. Eur J Med Genet 52: 404–408, 2009. Blough RI, Petrij F, Dauwerse JG, et al. Variation in microdeletions of the cyclic AMP-responsive element-binding protein gene at chromosome band 16p13.3 in the Rubinstein-Taybi syndrome. Am J Med Genet 90: 29–34, 2000. Blouin JL, Binkert F, Antonarakis SE. Biparental inheritance of chromosome 21 polymorphic markers indicates that some Robertsonian translocations t(21;21) occur postzygotically. Am J Med Genet 49: 363–368, 1994. Blumenthal AL, Allanson JE. Turner syndrome in a mother and daughter: r(X) and fertility. Clin Genet 52: 187–191, 1997. Bobabilla-Morales L, Corona-Rivera A , CoronaRivera JR , et al. Chromosome instability induced in vitro with mitomycin C in five Seckel syndrome patients. Am J Med Genet 123A : 148–152, 2003. Bobrow M, Barby T, Hajianpour A , Maxwell D, Yau SC. Fertility in a male with trisomy 21. J Med Genet 29: 141, 1992. Bochukova EG, Huang N, Keogh J, et al. Large, rare chromosomal deletions associated with severe early-onset obesity. Nature 463: 666–670, 2010. Bocian E, Mazurczak T, Stanczak H. Paracentric inversion inv(18)(q21.1q23) in a woman with recurrent spontaneous abortions. Am J Med Genet 35: 592–593, 1990. Bodri D, Guillén JJ, Schwenn K , Casadesus S, Vidal R , Coll O. Poor outcome in oocyte donation after elective transfer of a single cleavage-stage embryo in Turner syndrome patients. Fertil Steril 91: 1489–1492, 2009. Boehmer ALM, Brinkmann O, Bruggenwirth H, et al. Genotype versus phenotype in families with androgen insensitivity syndrome. J Clin Endocrinol Metab 86: 4151–4160, 2001. Bofinger MK , Needham DF, Saldana LR , Sosnowski JP, Blough RI. 45,X/46,X,r(Y) karyotype transmitted by father to son after intracytoplasmic sperm injection for oligospermia. A case report. J Reprod Med 44: 645–648, 1999. Boghosian-Sell L, Mewar R , Harrison W, et al. Molecular mapping of the Edwards syndrome phenotype to two noncontiguous regions on chromosome 18. Am J Hum Genet 55: 476–483, 1994. Bohers E, Sarafan-Vasseur N, Drouet A , et al. Gradual reduction of BUBR1 protein levels results in
premature sister-chromatid separation then in aneuploidy. Hum Genet 124: 473–478, 2008. Boivin J, Appleton TC, Baetens P, et al. Guidelines for counselling in infertility: outline version. Hum Reprod 16: 1301–1304, 2001. Bolor H, Mori T, Nishiyama S, et al. Mutations of the SYCP3 gene in women with recurrent pregnancy loss. Am J Hum Genet 84: 14–20, 2009. Bolton PF, Dennis NR , Browne CE, et al. The phenotypic manifestations of interstitial duplications of proximal 15q with special reference to the autistic spectrum disorders. Am J Med Genet 105: 675–685, 2001. Bonaglia MC, Ciccone R , Gimelli G, et al. Detailed phenotype-genotype study in five patients with chromosome 6q16 deletion: narrowing the critical region for Prader-Willi-like phenotype. Eur J Hum Genet 16: 1443–1449, 2008. Bonaglia MC, Giorda R , Carrozzo R , et al. 20-Mb duplication of chromosome 9p in a girl with minimal physical findings and normal IQ: narrowing of the 9p duplication critical region to 6 Mb. Am J Med Genet 112: 154–159, 2002. Bonaglia MC, Giorda R , Mani E, et al. Identification of a recurrent breakpoint within the SHANK3 gene in the 22q13.3 deletion syndrome. J Med Genet 43: 822–828, 2006. Bonaglia MC, Marelli S, Novara F, et al. Genotypephenotype relationship in three cases with overlapping 19p13.12 microdeletions. Eur J Hum Genet 18: 1302–1309, 2010. Bonduelle M, Van Assche E, Joris H, et al. Prenatal testing in ICSI pregnancies: incidence of chromosomal anomalies in 1586 karyotypes and relation to sperm parameters. Hum Reprod 17: 2600–2614, 2002. Bonnet C, Zix C, Grégoire MJ, et al. Characterization of mosaic supernumerary ring chromosomes by array-CGH: segmental aneusomy for proximal 4q in a child with tall stature and obesity. Am J Med Genet 140: 233–237, 2006. Boonen SE, Hoffmann AL, Donnai D, Tümer Z, Ravn K. Diploid/triploid mosaicism: A rare event or an under-diagnosed syndrome? Eur J Med Genet 54: 374–375, 2011. Boormans EMA , Birnie E, Oepkes D, et al. The impact of rapid aneuploidy detection (RAD) in addition to karyotyping versus karyotyping on maternal quality of life. Prenat Diagn 30: 425–433, 2010. Borgström B, Hreinsson J, Rasmussen C, et al. Fertility preservation in girls with turner syndrome: prognostic signs of the presence of ovarian follicles. J Clin Endocrinol Metab 94: 74–80, 2009.
Borie C, Léger J, Dupuy O, et al. Translocation (Y;22) resulting in the loss of SHOX and isolated short stature. Am J Med Genet 125A : 186–190, 2004. Bos AP, Broers CJ, Hazebroek FW, et al. Avoidance of emergency surgery in newborn infants with trisomy 18. Lancet 339: 913–915, 1992. Bottani A , Robinson WP, DeLozier-Blanchet CD, et al. Angelman syndrome due to paternal uniparental disomy of chromosome 15: a milder phenotype? Am J Med Genet 51: 35–40, 1994. Botto LD, Mulinare J, Yang Q, Liu Y, Erickson JD. Autosomal trisomy and maternal use of multivitamin supplements. Am J Med Genet 125A : 113–116, 2004. Boudreau EA , Johnson KP, Jackman AR , et al. Review of disrupted sleep patterns in Smith-Magenis syndrome and normal melatonin secretion in a patient with an atypical interstitial 17p11.2 deletion. Am J Med Genet 149A : 1382–1391, 2009. Boué A , Gallano P. A collaborative study of the segregation of inherited chromosome structural rearrangements in 1356 prenatal diagnoses. Prenat Diagn 4 Spec No: 45–67, 1984. Bound JP, Francis BJ, Harvey PW. Down’s syndrome: prevalence and ionising radiation in an area of north west England 1957–91. J Epidemiol Community Health 49: 164–170, 1995. Bourgeois JA , Coffey SM, Rivera SM, et al. A review of fragile X premutation disorders: expanding the psychiatric perspective. J Clin Psychiatry 70: 852–862, 2009. Bourthoumieu S, Esclaire F, Terro F, et al. “Cri-duchat” syndrome in a patient born to a mother with a paracentric inversion of chromosome 5q. Ann Génét 46: 483–486, 2003. Bovie C, Holden ST, Schroer A , Smith E, Trump D, Raymond FL. Neurofibromatosis 2 in a patient with a de novo balanced reciprocal translocation 46,X,t(X;22)(p11.2;q11.2). J Med Genet 40: 682–684, 2003. Bowen P, Fitzgerald PH, Gardner RJM, Biederman B, Veale AMO. Duplication 8q syndrome due to familial chromosome ins(10;8)(q21;q212q22). Am J Med Genet 14: 635–646, 1983. Bowser-Riley S, Buckton KE, Ratcliffe SG, Syme J. Inheritance of a ring 14 chromosome. J Med Genet 18: 209–213, 1981. Bowser-Riley SM, Griffiths MJ, Creasy MR , et al. Are double translocations double trouble? J Med Genet 25: 326–331, 1988. Boyd LJ, Livingston JS, Brown MG, et al. Meiotic exchange event within the stalk region of an inverted chromosome 22 results in a recombinant chromosome with duplication of the distal long arm. Am J Med Genet 138: 355–360, 2005.
REFERENCES
•
523
Boyd PA , Loane M, Garne E, Khoshnood B, Dolk H. Sex chromosome trisomies in Europe: prevalence, prenatal detection and outcome of pregnancy. Eur J Hum Genet 19: 231–234, 2011. Boyle J, Sangha K , Dill F, Robinson WP, Yong SL. Grandmaternal origin of an isochromosome 18p present in two maternal half-sisters. Am J Med Genet 101: 65–69, 2001. Braddock SR , Henley KM, Potter KL, Nguyen HG, Huang TH. Tertiary trisomy due to a reciprocal translocation of chromosomes 5 and 21 in a four-generation family. Am J Med Genet 92: 311–317, 2000. Brady AF, Waters CS, Pocha MJ, Brueton LA . Chronic myelomonocytic leukaemia in a child with constitutional partial trisomy 8 mosaicism. Clin Genet 58: 142–146, 2000. Brady AN, May KM, Fernhoff PM. Mosaic trisomy 4: Long-term outcome on the first reported liveborn. Am J Med Genet 132: 411–413, 2005. Brahams D. House of Lords upholds decision to sterilise 17-year-old mentally handicapped girl. Lancet 1: 1099–1100, 1987. Brambati B, Tului L, Camurri L, Guercilena S. Early second trimester (13 to 20 weeks) transabdominal chorionic villus sampling (TA-CVS): a safe and alternative method for both high and low risk populations. Prenat Diagn 22: 907–913, 2002. Brandt CA , Lüdecke HJ, Hindkjær J, et al. A de nevo complex t(7;13;8) translocation with a deletion in the TRPS gene region. Hum Genet 100: 334–338, 1997. Braun A , Kammerer S, Cleve H, Löhrs U, Schwarz HP, Kuhnle U. True hermaphroditism in a 46,XY individual, caused by a postzygotic somatic point mutation in the male gonadal sex-determining locus (SRY): molecular genetics and histological findings in a sporadic case. Am J Hum Genet 52: 578–585, 1993. Braun-Falco M, Schempp W, Nevinny-StickelHinzpeter C, Köhn FM. Azoospermia due to a unique de novo balanced reciprocal translocation (Y;1) (q12;q25). J Androl 28: 647–651, 2007. Breathnach FM, Malone FD, Lambert-Messerlian G, et al. First- and second-trimester screening: detection of aneuploidies other than Down syndrome. Obstet Gynecol 110: 651–657, 2007. Brender JD, Zhan FB, Langlois PH, Suarez L, Scheuerle A . Residential proximity to waste sites and industrial facilities and chromosomal anomalies in offspring. Int J Hyg Environ Health 211: 50–58, 2008. Brewer C, Holloway S, Zawalnyski P, Schinzel A , FitzPatrick D. A chromosomal deletion map of
524
•
REFERENCES
human malformations. Am J Hum Genet 63: 1153–1159, 1998. Brewer C, Holloway S, Zawalnyski P, Schinzel A , FitzPatrick D. A chromosomal duplication map of malformations: regions of suspected haplo- and triplolethality—and tolerance of segmental aneuploidy—in humans. Am J Hum Genet 64: 1702–1708, 1999. Brewer CM, Grace E, Stark GD, Gregory DW, Howell RT, Fitzpatrick DR . Genomic instability associated with limb defects: case report and review of the literature. Clin Dysmorphol 6: 99–109, 1997. Brewer CM, Holloway SH, Stone DH, Carothers AD, FitzPatrick DR . Survival in trisomy 13 and trisomy 18 cases ascertained from population based registers. J Med Genet 39: e54, 2002. Briault S, Odent S, Lucas J, et al. Paracentric inversion of the X chromosome [inv(X)(q12q28)] in familial FG syndrome. Am J Med Genet 86: 112–114, 1999. Brinkmann AO. Molecular basis of androgen insensitivity. Mol Cell Endocrinol 179: 105–109, 2001. Brisset S, Aboura A , Audibert F, et al. Discordant prenatal diagnosis of trisomy 21 due to mosaic structural rearrangements of chromosome 21. Prenat Diagn 23: 461–469, 2003. Brisset S, Izard V, Misrahi M, et al. Cytogenetic, molecular and testicular tissue studies in an infertile 45,X male carrying an unbalanced (Y;22) translocation: case report. Hum Reprod 20: 2168–2172, 2005. Britt DW, Risinger ST, Miller V, Mans MK , Krivchenia EL, Evans MI. Determinants of parental decisions after the prenatal diagnosis of down syndrome: bringing in context. Am J Med Genet 93: 410–416, 2000. Brizot ML, Schultz R , Patroni LT, Lopes LM, Armbruster-Moraes E, Zugaib M. Trisomy 10: ultrasound features and natural history after first trimester diagnosis. Prenat Diagn 21: 672–675, 2001. Brock DW. The non-identity problem and genetic harms—the case of wrongful handicaps. Bioethics 9: 269–275, 1995. Brockmann K , Böhm R , Bürger J. Exceptionally mild Angelman syndrome phenotype associated with an incomplete imprinting defect. J Med Genet 39: e51, 2002. Brøndum-Nielsen K , Mikkelsen M. A 10-year survey, 1980–1990, of prenatally diagnosed small supernumerary marker chromosomes, identified by FISH analysis. Outcome and follow-up of 14 cases diagnosed in a series of 12,699 prenatal samples. Prenat Diagn 15: 615–619, 1995.
Brouwer JR , Willemsen R , Oostra BA . The FMR1 gene and fragile X-associated tremor/ataxia syndrome. Am J Med Genet 150B: 782–798, 2009. Brown GM, Leversha M, Hultén M, Ferguson-Smith MA , Affara NA , Furlong RA . Genetic analysis of meiotic recombination in humans by use of sperm typing: reduced recombination within a heterozygous paracentric inversion of chromosome 9q32-q34.3. Am J Hum Genet 62: 1484–1492, 1998. Brown KK , Reiss JA , Crow K , et al. Deletion of an enhancer near DLX5 and DLX6 in a family with hearing loss, craniofacial defects, and an inv(7) (q21.3q35). Hum Genet 127: 19–31, 2010. Brown LY, Alonso ML, Yu J, Warburton D, Brown S. Prenatal diagnosis of a familial Xq deletion in a female fetus: a case report. Prenat Diagn 21: 27–30, 2001. Brown S, Higgins S, Hutchinson R , Bain S, Moore L, Haan E. A case of mosaicism for trisomy of chromosome 5 detected at amniocentesis. Twin Res Hum Genet 12: 210, 2009. Bruce S, Hannula-Jouppi K , Puoskari M, et al. Submicroscopic genomic alterations in SilverRussell syndrome and Silver-Russell-like patients. J Med Genet 47: 816–822, 2010. Brun JL, Gangbo F, Wen ZQ, et al. Prenatal diagnosis and management of sex chromosome aneuploidy: a report on 98 cases. Prenat Diagn 24: 213–218, 2004. Brun JL, Mangione R , Gangbo F, et al. Feasibility, accuracy and safety of chorionic villus sampling: a report of 10741 cases. Prenat Diagn 23: 295–301, 2003. Brunberg JA , Jacquemont S, Hagerman RJ, et al. Fragile X premutation carriers: characteristic MR imaging findings of adult male patients with progressive cerebellar and cognitive dysfunction. Am J Neuroradiol 23: 1757–1766, 2002. Brunet A , Gabau E, Perich RM, et al. Microdeletion and microduplication 22q11.2 screening in 295 patients with clinical features of DiGeorge/ Velocardiofacial syndrome. Am J Med Genet 140: 2426–2432, 2006. Brunetti-Pierri N, Berg JS, Scaglia F, et al. Recurrent reciprocal 1q21.1 deletions and duplications associated with microcephaly or macrocephaly and developmental and behavioral abnormalities. Nat Genet 40: 1466–1471, 2008. Brunetti-Pierri N, Paciorkowski AR , Ciccone R , et al. Duplications of FOXG1 in 14q12 are associated with developmental epilepsy, mental retardation, and severe speech impairment. Eur J Hum Genet 19: 102–107, 2011.
Bruns D. Presenting physical characteristics, medical conditions and developmental status of long-term survivors with trisomy 9 mosaicism. Am J Med Genet 155A :1033–1039, 2011. Bruyère H, Robertson G, Wilson RD, Langlois S. Risk of mosaicism and uniparental disomy associated with the prenatal diagnosis of non homologous Robertsonian translocation carrier. Am J Hum Genet 69: 208, 2001. Bruyère H, Rupps R , Kuchinka BD, Friedman JM, Robinson WP. Recurrent trisomy 21 in a couple with a child presenting trisomy 21 mosaicism and maternal uniparental disomy for chromosome 21 in the euploid cell line. Am J Med Genet 94: 35–41, 2000. Bruyère H, Speevak MD, Winsor EJ, et al. Isodicentric Yp: prenatal diagnosis and outcome in 12 cases. Prenat Diagn 26: 324–329, 2006. Bryan J, Peters M, Pritchard G, Healey S, Payton D. A second case of intrauterine growth retardation and primary hypospadias associated with a trisomy 22 placenta but with biparental inheritance of chromosome 22 in the fetus. Prenat Diagn 22: 137–140, 2002. Bryant LD, Murray J, Green JM, Hewison J, Sehmi I, Ellis A . Descriptive information about Down syndrome: a content analysis of serum screening leaflets. Prenat Diagn 21: 1057–1063, 2001. Buckton KE, Newton MS, Collyer S, et al. Phenotypically normal individuals with an inversion (X) (p22q13) and the recombinant (X), dup q. Ann Hum Genet 45: 159–168, 1981. Bugge M, Bruun-Petersen G, Brøndum-Nielsen K , et al. Disease associated balanced chromosome rearrangements: a resource for large scale genotype-phenotype delineation in man. J Med Genet 37: 858–865, 2000. Bugge M, Collins A , Hertz JM, et al. Non-disjunction of chromosome 13. Hum Mol Genet 16: 2004–2010, 2007. Bugge M, Collins A , Petersen MB, et al. Nondisjunction of chromosome 18. Hum Mol Genet 7: 661–669, 1998. Bugge M, deLozier-Blanchet C, Bak M, et al. Trisomy 13 due to rea(13q;13q) is caused by i(13) and not rob(13;13)(q10;q10) in the majority of cases. Am J Med Genet 132A : 310–313, 2005. Bui TH, Vetro A , Zuffardi O, Shaffer LG. Current controversies in prenatal diagnosis 3: is conventional chromosome analysis necessary in the post-array CGH era? Prenat Diagn 31: 235–243, 2011. Buiting K , Dittrich B, Gross S, et al. Sporadic imprinting defects in Prader-Willi syndrome and Angelman syndrome: implications for
REFERENCES
•
525
imprint-switch models, genetic counseling, and prenatal diagnosis. Am J Hum Genet 63: 170–180, 1998. Buiting K , Gross S, Lich C, Gillessen-Kaesbach G, el-Maarri O, Horsthemke B. Epimutations in Prader-Willi and Angelman syndromes: a molecular study of 136 patients with an imprinting defect. Am J Hum Genet 72: 571–577, 2003. Bukovsky A , Caudle MR , Svetlikova M, Upadhyaya NB. Origin of germ cells and formation of new primary follicles in adult human ovaries. Reprod Biol Endocrinol 2: 20, 2004. Bukvic N, Carri VD, Di Cosola ML, et al. Familial X;Y translocation with distinct phenotypic consequences: characterization using FISH and array CGH. Am J Med Genet 152A : 1730–1734, 2010. Bukvic N, Gentile M, Susca F, et al. Sex chromosome loss, micronuclei, sister chromatid exchange and aging: a study including 16 centenarians. Mutat Res 498: 159–167, 2001. Burke LW, Wiley JE, Glenn CC, et al. Familial cryptic translocation resulting in Angelman syndrome: implications for imprinting or location of the Angelman gene? Am J Hum Genet 58: 777–784, 1996. Burn J, Baraitser M, Butler LJ. An avoidable recurrence of cri du chat syndrome in the next generation. Br Med J 287: 1287–1288, 1983. Burnett AC, Reutens DC, Wood AG. Social cognition in Turner’s Syndrome. J Clin Neurosci 17: 283–286, 2010. Burns JP, Koduru PR , Alonso ML, Chaganti RS. Analysis of meiotic segregation in a man heterozygous for two reciprocal translocations using the hamster in vitro penetration system. Am J Hum Genet 38: 954–964, 1986. Burnside RD, Lose EJ, Dominguez MG, et al. Molecular cytogenetic characterization of two cases with constitutional distal 11q duplication/ triplication. Am J Med Genet 149A : 1516–1522, 2009. Burrello N, Calogero AE, De Palma A , et al. Chromosome analysis of epididymal and testicular spermatozoa in patients with azoospermia. Eur J Hum Genet 10: 362–366, 2002. Busche A , Graul-Neumann LM, Zweier C, Rauch A , Klopocki E, Horn D. Microdeletions of chromosome 7p21, including TWIST1, associated with significant microcephaly, facial dysmorphism, and short stature. Eur J Med Genet 54: 256–261, 2011. Buysse K , Delle Chiaie B, Van Coster R , et al. Challenges for CNV interpretation in clinical molecular karyotyping: lessons learned from a
526
•
REFERENCES
1001 sample experience. Eur J Med Genet 52: 398–403, 2009. Byrne J. Long-term genetic and reproductive effects of ionizing radiation and chemotherapeutic agents on cancer patients and their offspring. Teratology 59: 210–215, 1999. Byrne J, Mulvihill JJ, Myers MH, et al. Effects of treatment on fertility in long-term survivors of childhood or adolescent cancer. N Engl J Med 317: 1315–1321, 1987. Byrne J, Rasmussen SA , Steinhorn SC, et al. Genetic disease in offspring of long-term survivors of childhood and adolescent cancer. Am J Hum Genet 62: 45–52, 1998. Cabrejo L, Guyant-Maréchal L, Laquerrière A , et al. Phenotype associated with APP duplication in five families. Brain 129: 2966–2976, 2006. Cacciagli P, Haddad MR , Mignon-Ravix C, et al. Disruption of the ATP8A2 gene in a patient with a t(10;13) de novo balanced translocation and a severe neurological phenotype. Eur J Hum Genet 18: 1360–1363, 2010. Caer E, Perrin A , Douet-Guilbert N, Amice V, De Braekeleer M, Morel F. Differing mechanisms of meiotic segregation in spermatozoa from three carriers of a pericentric inversion of chromosome 8. Fertil Steril 89: 1637–1640, 2008. Cai T, Yu P, Tagle DA , Lu D, Chen Y, Xia J. A de novo complex chromosomal rearrangement with a translocation 7;9 and 8q insertion in a male carrier with no infertility. Hum Reprod 16: 59–62, 2001. Calabrese G, Fischetto R , Stuppia L, et al. X/Y translocation in a family with Leri-Weill dyschondrosteosis. Hum Genet 105: 367–368, 1999. Callen DF, Eyre H, Fang YY, et al. Origins of accessory small ring marker chromosomes derived from chromosome 1. J Med Genet 36: 847–853, 1999. Callen DF, Eyre HJ, Ringenbergs ML. A dicentric variant of chromosome 6: characterization by use of in situ hybridisation with the biotinylated probe p308. Clin Genet 37: 81–83, 1990. Callen DF, Eyre HJ, Ringenbergs ML, Freemantle CJ, Woodroffe P, Haan EA . Chromosomal origin of small ring marker chromosomes in man: characterization by molecular genetics. Am J Hum Genet 48: 769–782, 1991. Callen DF, Eyre H, Yip MY, Freemantle J, Haan EA . Molecular cytogenetic and clinical studies of 42 patients with marker chromosomes. Am J Med Genet 43: 709–715, 1992. Callen DF, Sutherland GR . Normal female carrier and affected male half-sibs with t(X;5)(q13;p15). Location of a gene determining male genital development. Clin Genet 30: 59–62, 1986.
Callen DF, Sutherland GR , Carter RF. A fertile man with tdic(Y;22): how a stable neo-X1X2Y sex-determining mechanism could evolve in man. Am J Med Genet Suppl 3: 151–155, 1987. Callen DF, Woollatt E, Sutherland GR . Paracentric inversions in man. Clin Genet 28: 87–92, 1985. Callier P, Faivre L, Pigeonnat S, et al. Contribution of array CGH in prognosis and genetic counselling of prenatally diagnosed supernumerary ring chromosome 20. Prenat Diagn 29: 1002–1005, 2009. Calounova G, Novotna D, Simandlova M, et al. Prader-Willi syndrome due to uniparental disomy in a patient with a balanced chromosomal translocation. Neuro Endocrinol Lett 27: 579–585, 2006. Calvari V, Bertini V, De Grandi A , et al. A new submicroscopic deletion that refines the 9p region for sex reversal. Genomics 65: 203–212, 2000. Cammarata M, Di Simone P, Graziano L, Giuffre M, Corsello G, Garofalo G. Rare sex chromosome aneuploidies in humans: report of six patients with 48,XXYY, 49,XXXXY, and 48,XXXX karyotypes. Am J Med Genet 85: 86–87, 1999. Canevini MP, Sgro V, Zuffardi O, et al. Chromosome 20 ring: a chromosomal disorder associated with a particular electroclinical pattern. Epilepsia 39: 942–951, 1998. Cans C, Cohen O, Lavergne C, Mermet MA , Demongeot J, Jalbert P. Logistic regression model to estimate the risk of unbalanced offspring in reciprocal translocations. Hum Genet 92: 598–604, 1993. Canto P, Söderlund D, Reyes E, Méndez JP. Mutations in the Desert hedgehog (DHH) gene in patients with 46,XY complete pure gonadal dysgenesis. J Clin Endocrinol Metab 89: 4480–4483, 2004. Canto P, Vilchis F, Söderlund D, Reyes E, Méndez JP. A heterozygous mutation in the desert hedgehog gene in patients with mixed gonadal dysgenesis. Mol Hum Reprod 11: 833–836, 2005. Carbone JF, Tuuli MG, Dicke JM, Macones GA , Odibo AO. Revisiting the risk for aneuploidy in fetuses with isolated pyelectasis. Prenat Diagn 31: 566–570, 2011. Carbonell P, López I, Gabarrón J, et al. FRAXE mutation analysis in three Spanish families. Am J Med Genet 64: 434–440, 1996. Cardoso C, Leventer RJ, Dowling JJ, et al. Clinical and molecular basis of classical lissencephaly: Mutations in the LIS1 gene (PAFAH1B1). Hum Mutat 19: 4–15, 2002. Cardoso C, Leventer RJ, Ward HL, et al. Refinement of a 400-kb critical region allows genotypic differentiation between isolated lissencephaly,
Miller-Dieker syndrome, and other phenotypes secondary to deletions of 17p13.3. Am J Hum Genet 72: 918–930, 2003. Cardoso LCA , Moraes L, Camilo MJE, et al. Cytogenetic and molecular studies of an X;21 translocation previously diagnosed as complete monosomy 21. Eur J Med Genet 51: 588–597, 2008. Carella M, Spreafico F, Palumbo O, et al. Constitutional ring chromosome 11 mosaicism in a Wilms tumor patient: cytogenetic, molecular and clinico-pathological studies. Am J Med Genet 152A : 1756–1763, 2010. Carey JC. Trisomy 18 and 13 syndromes. In: Cassidy SB, Allanson JE, eds. Management of Genetic Syndromes. New York: Wiley-Liss, 2001: 555–568. Carothers AD, Buckton KE, Collyer S, et al. The effect of variant chromosomes on reproductive fitness in man. Clin Genet 21: 280–289, 1982. Carothers AD, Castilla EE, Dutra MG, Hook EB. Search for ethnic, geographic, and other factors in the epidemiology of Down syndrome in South America: analysis of data from the ECLAMC project, 1967–1997. Am J Med Genet 103: 149–156, 2001. Carothers AD, Hecht CA , Hook EB. International variation in reported livebirth prevalence rates of Down syndrome, adjusted for maternal age. J Med Genet 36: 386–393, 1999. Carp H, Toder V, Aviram A , Daniely M, Mashiach S, Barkai G. Karyotype of the abortus in recurrent miscarriage. Fertil Steril 75: 678–682, 2001. Carter MT, St Pierre SA , Zackai EH, Emanuel BS, Boycott KM. Phenotypic delineation of Emanuel syndrome (supernumerary derivative 22 syndrome): clinical features of 63 individuals. Am J Med Genet 149A : 1712–1721, 2009. Casasbuenas A , Wong AE, Sepulveda W. Firsttrimester nasal bone length in a normal Latin American population. Prenat Diagn 29: 108–112, 2009. Casper AM, Durkin SG, Arlt MF, Glover TW. Chromosomal instability at common fragile sites in Seckel syndrome. Am J Hum Genet 75: 654–660, 2004. Cassidy SB, Allanson JE. Management of Genetic Syndromes. New York: Wiley-Liss, 2001. Cassidy SB, Forsythe M, Heeger S, et al. Comparison of phenotype between patients with Prader-Willi syndrome due to deletion 15q and uniparental disomy 15. Am J Med Genet 68: 433–440, 1997. Cassidy SB, Lai LW, Erickson RP, et al. Trisomy 15 with loss of the paternal 15 as a cause of PraderWilli syndrome due to maternal disomy. Am J Hum Genet 51: 701–708, 1992.
REFERENCES
•
527
Cassidy SB, Whitworth T, Sanders D, Lorber CA , Engel E. Five month extrauterine survival in a female triploid (69,XXX) child. Ann Génét 20: 277–279, 1977. Castella M, Pujol R , Callén E, et al. Chromosome fragility in patients with Fanconi anaemia: diagnostic implications and clinical impact. J Med Genet 48: 242–250, 2011. Castellví-Bel S, Fernández-Burriel M, Rifé M, et al. Detection of the fragile X syndrome protein for the evaluation of FMR1 intermediate alleles. Hum Genet 107: 195–196, 2000. Castellví-Bel S, Milà M, Soler A , et al. Prenatal diagnosis of fragile X syndrome: (CGG)n expansion and methylation of chorionic villus samples. Prenat Diagn 15: 801–807, 1995. Castellví-Bel S, Sánchez A , Badenas C, et al. Singlestrand conformation polymorphism analysis in the FMR1 gene. Am J Med Genet 84: 262–265, 1999. Castiglia D, Castori M, Pisaneschi E, et al. Trisomic rescue causing reduction to homozygosity for a novel ABCA12 mutation in harlequin ichthyosis. Clin Genet 76: 392–397, 2009. Castorina P, Selicorni A , Bedeschi F, Dalpra L, Larizza L. Genotype-phenotype correlation in two sets of monozygotic twins with Williams syndrome. Am J Med Genet 69: 107–111, 1997. Catelani ALPM, Krepischi ACV, Kim CA , et al. Chromosome imbalances in syndromic hearing loss. Clin Genet 76: 458–464, 2009. Causio F, Fischetto R , Leonetti T, Schonauer LM. Ovarian stimulation in a woman with premature ovarian failure and X-autosome translocation. A case report. J Reprod Med 45: 235–239, 2000. Causio F, Fischetto R , Schonauer LM, Leonetti T. Intracytoplasmic sperm injection in infertile patients with structural cytogenetic abnormalities. J Reprod Med 44: 859–864, 1999. Celle L, Lee L, Rintoul N, et al. Duplication of chromosome region 4q28.3-qter in monozygotic twins with discordant phenotypes. Am J Med Genet 94: 125–140, 2000. Cerruti Mainardi PC, Perfumo C, Cali A , et al. Clinical and molecular characterisation of 80 patients with 5p deletion: genotype-phenotype correlation. J Med Genet 38: 151–158, 2001. Chakrabarti L, Best TK , Cramer NP, et al. Olig1 and Olig2 triplication causes developmental brain defects in Down syndrome. Nature Neurosci 13: 927–934, 2010. Chan CTJ, Clayton-Smith J, Cheng XJ, et al. Molecular mechanisms in Angelman syndrome: a survey of 93 patients. J Med Genet 30: 895–902, 1993.
528
•
REFERENCES
Chan Y, Silverman N, Jackson L, Wapner R , Wallerstein R . Maternal uniparental disomy of chromosome 16 and body stalk anomaly. Am J Med Genet 94: 284–286, 2000. Chandley AC. Asymmetry in chromosome pairing: a major factor in de novo mutation and the production of genetic disease in man. J Med Genet 26: 546–552, 1989. Chang HJ, Clark RD, Bachman H. The phenotype of 45,X/46,XY mosaicism: an analysis of 92 prenatally diagnosed cases. Am J Hum Genet 46: 156–167, 1990. Chang LW, Chen PY, Kuo PL, Chang FM. Prenatal diagnosis of a fetus with megacystis and monosomy 21. Prenat Diagn 21: 512–513, 2001. Chang PL, Sauer MV, Brown S. Y chromosome microdeletion in a father and his four infertile sons. Hum Reprod 14: 2689–2694, 1999. Chatzimeletiou K , Makrydimas G, Sotiriadis A , Paraskevaidis E, Nicolaides KH. Aneuploidy screening in coelomic samples using fluorescence in situ hybridisation (FISH). Prenat Diagn 25: 919–926, 2005. Check JH, Katsoff B, Summers-Chase D, Breitbart J. A case report supporting the concept that some women have a predisposition for maternal meiosis errors resulting in digyny. Clin Exp Obstet Gynecol 36: 133–134, 2009. Cheffins T, Chan A , Haan EA , et al. The impact of maternal serum screening on the birth prevalence of Down’s syndrome and the use of amniocentesis and chorionic villus sampling in South Australia. Br J Obstet Gynaecol 107: 1453–1459, 2000. Chen CL, Gilbert TJ, Daling JR . Maternal smoking and Down syndrome: the confounding effect of maternal age. Am J Epidemiol 149: 442–446, 1999a. Chen CP, Chern SR , Chang TY, et al. Prenatal diagnosis of de novo proximal interstitial deletion of 9q and review of the literature of uncommon aneuploidies associated with increased nuchal translucency. Prenat Diagn 25: 383–389, 2005a. Chen CP, Chern SR , Chen LF, Chen WL, Wang W. Prenatal diagnosis of low-level mosaic trisomy 7 by amniocentesis. Prenat Diagn 25: 1067–1069, 2005b. Chen CP, Chern SR , Lee CC, Chang TY, Wang W, Tzen CY. Clinical, cytogenetic, and molecular findings of prenatally diagnosed mosaic trisomy 4. Prenat Diagn 24: 38–44, 2004a. Chen CP, Chern SR , Lee CC, Chen WL, Wang W. Prenatal diagnosis of interstitially satellited 6p. Prenat Diagn 24: 430–433, 2004b. Chen CP, Chern SR , Lee CC, et al. Prenatal diagnosis of de novo t(2;18;14)(q33.1;q12.2;q31.2), dup(5)
(q34q34), del(7)(p21.1p21.1), and del(10) (q25.3q25.3) and a review of the prenatally ascertained de novo apparently balanced complex and multiple chromosomal rearrangements. Prenat Diagn 26: 138–146, 2006a. Chen CP, Chern SR , Lee CC, Town DD. Isochromosome 18q in a fetus with congenital megacystis, intra-uterine growth retardation and cloacal dysgenesis sequence. Prenat Diagn 18: 1068–1074, 1998. Chen CP, Chern SR , Lee PY, Town DD, Wang W. Prenatal diagnosis of low-level mosaic trisomy 6 by amniocentesis. Prenat Diagn 26:1093–1096, 2006b. Chen CP, Chern SR , Sheu JC, et al. Prenatal diagnosis, sonographic findings and molecular genetic analysis of a 46,XX/46,XY true hermaphrodite chimera. Prenat Diagn 25: 502–506, 2005c. Chen CP, Chern SR , Tsai FJ, et al. Down syndrome due to unbalanced homologous acrocentric rearrangements and its recurrence in subsequent pregnancies: prenatal diagnosis by amniocentesis. Taiwan J Obstet Gynecol 48: 403–407, 2009. Chen CP, Chern SR , Town DD, Wang W, Liao YW. Fetoplacental and fetoamniotic chromosomal discrepancies in prenatally detected mosaic trisomy 9. Prenat Diagn 23: 1019–1021, 2003a. Chen CP, Chern SR , Wang TY, Wang W, Wang KL, Jeng CJ. Androgen receptor gene mutations in 46,XY females with germ cell tumours. Hum Reprod 14: 664–670, 1999b. Chen CP, Devriendt K , Chern SR , Lee CC, Wang W, Lin SP. Prenatal diagnosis of inherited satellited non-acrocentric chromosomes. Prenat Diagn 20: 384–389, 2000. Chen CP, Hsu CY, Tzen CY, et al. Prenatal diagnosis of mosaic ring chromosome 4. Prenat Diagn 27:485–487, 2007a. Chen CP, Hwu YM, Yeh LF, Chern SR , Lee CC, Wang W. Successful triplet pregnancy and delivery after oocyte donation in an infertile female with chromosome mosaicism for monosomy X, partial trisomy X, and terminal Xp deletion. Fertil Steril 79: 1231–1233, 2003b. Chen CP, Lee CC, Chen WL, Wang W, Tzen CY. Prenatal diagnosis of premature centromere division-related mosaic variegated aneuploidy. Prenat Diagn 24: 19–25, 2004c. Chen CP, Lee CC, Town DD, et al. Detection of euchromatic variants and unusual C band heterochromatin variants at genetic amniocentesis. Genet Couns 17: 91–95, 2006c. Chen CP, Lee CC, Wang W. Prenatal diagnosis of complete trisomy 16q in two consecutive pregnancies. Prenat Diagn 24: 928–929, 2004d.
Chen CP, Lin CL, Chen LL, Lee CC, Wang W. Prenatal diagnosis of mosaic ring chromosome 9. Prenat Diagn 26:870–871, 2006d. Chen CP, Lin SP, Lin CC, et al. Prenatal diagnosis of low-level mosaicism for a small XIST-negative supernumerary ring X chromosome in a nondysmorphic male fetus. Prenat Diagn 26: 387–391, 2006e. Chen CP, Lin SP, Sheu JC, Wang W, Tzen CY. Neonatal outcome of a prenatally detected 46,XX/46,XY true hermaphrodite. Prenat Diagn 26: 185–186, 2006f. Chen CP, Lin SP, Tsai FJ, Wang TH, Chern SR , Wang W. Characterization of a de novo unbalanced Y; autosome translocation in a 45,X mentally retarded male and literature review. Fertil Steril 90: 1198 e11–18, 2008. Chen CP, Su YN, Lin CC, et al. Genetic counseling of prenatally detected unbalanced t(Y;15)(q12;p13). Genet Couns 18: 455–457, 2007b. Chen E, Choe MA , Loughman WD, et al. Recurrent adjacent-2 segregation of a familial t(14;21) (q11.2;q11.2): phenotypic comparison of two brothers and a paternal aunt inheriting the der(14). Am J Med Genet 132A : 164–170, 2005d. Chen H, Young R , Mu X , et al. Uniparental isodisomy resulting from 46,XX,i(1p),i(1q) in a woman with short stature, ptosis, micro/retrognathia, myopathy, deafness, and sterility. Am J Med Genet 82: 215–218, 1999c. Chen M, Yeh GP, Shih JC, Wang BT. Trisomy 13 mosaicism: study of serial cytogenetic changes in a case from early pregnancy to infancy. Prenat Diagn 24: 137–143, 2004e. Chen Y, Huang J, Liu P, Qiao J. Analysis of meiotic segregation patterns and interchromosomal effects in sperm from six males with Robertsonian translocations. J Assist Reprod Genet 24: 406–411, 2007c. Chenevix-Trench G, Spurdle AB, Gatei M, et al. Dominant negative ATM mutations in breast cancer families. J Natl Cancer Inst 94: 205–215, 2002. Cheng DH, Tan YQ, Di YF, Li LY, Lu GX . Crypt Y chromosome fragment resulting from an X;Y translocation in a patient with premature ovarian failure. Fertil Steril 92: 828 e3–6, 2009. Cheng EY, Chen YJ, Disteche CM, Gartler SM. Analysis of a paracentric inversion in human oocytes: nonhomologous pairing in pachytene. Hum Genet 105: 191–196, 1999. Chessa L, Piane M, Prudente S, et al. Molecular prenatal diagnosis of ataxia telangiectasia heterozygosity by direct mutational assays. Prenat Diagn 19: 542–545, 1999.
REFERENCES
•
529
Chettouh Z, Croquette MF, Delobel B, et al. Molecular mapping of 21 features associated with partial monosomy 21: involvement of the APP-SOD1 region. Am J Hum Genet 57: 62–71, 1995. Chia N, Bousfield L, James S, Nelson J. Report of a severely retarded child with a duplication 18q derived from a paternal paracentric inversion (18q): a note of warning. Bull Hum Genet Soc Australasia 6: 48, 1992. Chia N, Daniel A , Malafiej P, Clarke N, Ades L. A child with a deletion 2q37 found to be a recombinant of a cryptic pericentric inversion: implications for phenotype and risk of recurrence. Bull Hum Genet Soc Australasia 14: 29, 2001. Chia NL. A comprehensive set of idiograms representing all interpretive levels of resolution: ISCN (2009). Cytogenet Genome Res 125: 162–164, 2009. Chiesa J, Hoffet M, Rousseau O, et al. Pallister-Killian syndrome [i(12p)]: first pre-natal diagnosis using cordocentesis in the second trimester confirmed by in situ hybridization. Clin Genet 54: 294–302, 1998. Chisholm CA , Bray MJ, Karns LB. Successful pregnancy in a woman with Bloom syndrome. Am J Med Genet 102: 136–138, 2001. Chitayat D, Wyatt PR , Wilson RD, et al. Fragile X testing in obstetrics and gynaecology in Canada. J Obstet Gynaecol Can 30: 837–846, 2008. Chiu RWK , Akolekar R , Zheng YWL, et al. Non-invasive prenatal assessment of trisomy 21 by multiplexed maternal plasma DNA sequencing: large scale validity study. Brit Med J 342: c740, 2011. Chiu RWK , Cantor CR , Lo YMD. Non-invasive prenatal diagnosis by single molecule counting technologies. Trends Genet 25: 324–331, 2009. Chiurazzi P. Mental retardation: is naming the real issue? Am J Med Genet 155A : 974–975, 2011. Chonchaiya W, Nguyen DV, Au J, et al. Clinical involvement in daughters of men with fragile X-associated tremor ataxia syndrome. Clin Genet 78: 38–46, 2010. Chonchaiya W, Schneider A , Hagerman RJ. Fragile X: a family of disorders. Adv Pediatr 56: 165–186, 2009. Christ LA , Crowe CA , Micale MA , Conroy JM, Schwartz S. Chromosome breakage hotspots and delineation of the critical region for the 9p-deletion syndrome. Am J Hum Genet 65: 1387–1395, 1999. Christian SM, Koehn D, Pillay R , MacDougall A , Wilson RD. Parental decisions following prenatal
530
•
REFERENCES
diagnosis of sex chromosome aneuploidy: a trend over time. Prenat Diagn 20: 37–40, 2000. Chu C, Schwartz S, McPherson E. Paternal uniparental isodisomy for chromosome 14 in a patient with a normal 46,XY karyotype. Am J Med Genet 127A : 167–171, 2004. Chuang L, Wakui K , Sue WC, Su MH, Shaffer LG, Kuo PL. Interstitial deletion 11(p11.12p11.2) and analphoid marker formation results in inherited Potocki-Shaffer syndrome. Am J Med Genet 133A : 180–183, 2005. Chudley AE, Bauder F, Ray M, McAlpine PJ, Pena SDJ, Hamerton JL. Familial mental retardation in a family with an inherited chromosome rearrangement. J Med Genet 11: 353–366, 1974. Chudley AE, Zheng HZ, Pabello PD, Shia G, Wang HC. Familial supernumerary microchromosome mosaicism: phenotypic effects and an attempt at characterization. Am J Med Genet 16: 89–97, 1983. Clarke A . The genetic testing of children. Working Party of the Clinical Genetics Society (UK). J Med Genet 31: 785–797, 1994. Clarkson B, Pavenski K , Dupuis L, et al. Detecting rearrangements in children using subtelomeric FISH and SKY. Am J Med Genet 107: 267–274, 2002. Clayton EW. Removing the shadow of the law from the debate about genetic testing of children. Am J Med Genet 57: 630–634, 1995. Clayton-Smith J, Laan L. Angelman syndrome: a review of the clinical and genetic aspects. J Med Genet 40: 87–95, 2003. Clayton-Smith J, Walters S, Hobson E, et al. Xq28 duplication presenting with intestinal and bladder dysfunction and a distinctive facial appearance. Eur J Hum Genet 17: 434–443, 2009. Clement Wilson S, Susman M, Bain S, et al. Isochromosome 5p mosaicism at prenatal diagnosis: observations and outcomes in six cases at chorionic villus sampling and one at amniocentesis. Prenat Diagn 22: 681–685, 2002. Clouston HJ, Herbert M, Fenwick J, Murdoch AP, Wolstenholme J. Cytogenetic analysis of human blastocysts. Prenat Diagn 22: 1143–1152, 2002. Cobben JM, Verheij JBGM, Eisma WH, et al. Bilateral split hand/foot malformation and inv(7) (p22q21.3). J Med Genet 32: 375–378, 1995. Cockwell AE, Baker SJ, Connarty M, Moore IE, Crolla JA . Mosaic trisomy 6 and maternal uniparental disomy 6 in a 23-week gestation fetus with atrioventricular septal defect. Am J Med Genet 140: 624–627, 2006. Cockwell A , Hatchwell E, Crolla J. Interchange trisomy resulting from the unbalanced segregation
of a maternal (1;7) chromosome. Eur J Hum Genet 4(suppl 1): 123, 1996a. Cockwell AE, Jacobs PA , Beal SJ, Crolla JA . A study of cryptic terminal chromosome rearrangements in recurrent miscarriage couples detects unsuspected acrocentric pericentromeric abnormalities. Hum Genet 112: 298–302, 2003. Cockwell AE, Jacobs PA , Crolla JA . Distribution of the D15Z1 copy number polymorphism. Eur J Hum Genet 15: 441–445, 2007. Cockwell AE, James RS, Moore IE, Hatchwell E, Crolla JA . Clinical outcomes of adjacent 1 segregation in a familial translocation t(8;18) (p21.3;p11.23). J Med Genet 33: 515–517, 1996b. Codina-Pascual M, Oliver-Bonet M, Navarro J, et al. FISH characterization of a dicentric Yq (p11.32) isochromosome in an azoospermic male. Am J Med Genet 127A : 302–306, 2004. Cody JD, Heard PL, Crandall AC, et al. Narrowing critical regions and determining penetrance for selected 18q- phenotypes. Am J Med Genet 149A : 1421–1430, 2009. Coffee B, Keith K , Albizua I, et al. Incidence of fragile X syndrome by newborn screening for methylated FMR1 DNA . Am J Hum Genet 85: 503–514, 2009. Coffey SM, Cook K , Tartaglia N, et al. Expanded clinical phenotype of women with the FMR1 premutation. Am J Med Genet 146A : 1009–1016, 2008. Cohen MM, Frederick RW, Balkin NE, Simpson SJ. The identification of Y chromosome translocations following Distamycin A treatment. Clin Genet 19: 335–342, 1981. Cohen O, Cans C, Mermet MA , Demongeot J, Jalbert P. Viability thresholds for partial trisomies and monosomies. A study of 1,159 viable unbalanced reciprocal translocations. Hum Genet 93: 188–194, 1994. Cohen O, Mermet MA , Demongeot J. HC Forum: a web site based on an international human cytogenetic database. Nucleic Acids Res 29: 305–307, 2001. Cohen O, Simonet M, Cans C, et al. Human reciprocal translocations: a new computer system for genetic counseling. Ann Génét 35: 193–201, 1992. Coles K , Mackenzie M, Crolla J, et al. A complex rearrangement associated with sex reversal and the Wolf-Hirschhorn syndrome: a cytogenetic and molecular study. J Med Genet 29: 400–406, 1992. Collado F, Bombard A , Li V, Julliard K , Aptekar L, Weiner Z. Ethnic variation of fetal nasal bone length between 11–14 weeks’ gestation. Prenat Diagn 25: 690–692, 2005.
Colleaux L, Rio M, Heuertz S, et al. A novel automated strategy for screening cryptic telomeric rearrangements in children with idiopathic mental retardation. Eur J Hum Genet 9: 319–327, 2001. Collins GM, Kim DU, Logrono R , Rickert RR , Zablow A , Breen JL. Pure seminoma arising in androgen insensitivity syndrome (testicular feminization syndrome): a case report and review of the literature. Mod Pathol 6: 89–93, 1993. Collins VR , Webley C, Sheffield LJ, Halliday JL. Fetal outcome and maternal morbidity after early amniocentesis. Prenat Diagn 18: 767–772, 1998. Collinson MN, Fisher AM, Walker J, Currie J, Williams L, Roberts P. Inv(10)(p11.2q21.2), a variant chromosome. Hum Genet 101: 175–180, 1997. Collinson MN, Roberts SE, Crolla JA , Dennis NR . A familial balanced inverted insertion ins(15) (q15q13q11.2) producing Prader-Willi syndrome, Angelman syndrome and duplication of 15q11.2-q13 in a single family: importance of differentiation from a paracentric inversion. Am J Med Genet 126A : 27–32, 2004. Colls P, Blanco J, Martínez-Pasarell O, et al. Chromosome segregation in a man heterozygous for a pericentric inversion, inv(9)(p11q13), analyzed by using sperm karyotyping and two-color fluorescence in situ hybridization on sperm nuclei. Hum Genet 99: 761–765, 1997. Coman DJ, Gardner RJM. Deletions that reveal recessive genes. Eur J Hum Genet 15: 1103–1104, 2007. Coman D, Gardner RJM, Pertile MD, Kannu P. Trisomy 16 mosaicism at chorionic villus sampling and amniocentesis with a normal physical and intellectual outcome. Fetal Diagn Ther 28: 117–118, 2010a. Coman D, Yaplito-Lee J, La P, et al. Three Mendelian disorders (chronic granulomatous disease, retinitis pigmentosa, ornithine transcarbamylase deficiency) in a young woman with an X chromosome deletion, del(X)(p11.4p21.1). Mol Genet Metab 99: 329, 2010b. Concolino D, Iembo MA , Rossi E, et al. Familial pericentric inversion of chromosome 5 in a family with benign neonatal convulsions. J Med Genet 39: 214–216, 2002. Conlin LK , Kramer W, Hutchinson AL, et al. Molecular analysis of ring chromosome 20 syndrome reveals two distinct groups of patients. J Med Genet 48: 1–9, 2011. Conn CM, Cozzi J, Harper JC, Winston RM, Delhanty JD. Preimplantation genetic diagnosis for couples at high risk of Down syndrome
REFERENCES
•
531
pregnancy owing to parental translocation or mosaicism. J Med Genet 36: 45–50, 1999. Conn CM, Harper JC, Winston RM, Delhanty JD. Infertile couples with Robertsonian translocations: preimplantation genetic analysis of embryos reveals chaotic cleavage divisions. Hum Genet 102: 117–123, 1998. Connerton-Moyer KJ, Nicholls RD, Schwartz S, et al. Unexpected familial recurrence in Angelman syndrome. Am J Med Genet 70: 253–260, 1997. Coonen E, Martini E, Dumoulin JC, et al. Preimplantation genetic diagnosis of a reciprocal translocation t(3;11)(q27.3;q24.3) in siblings. Mol Hum Reprod 6: 199–206, 2000. Cooper GM, Nickerson DA , Eichler EE. Mutational and selective effects on copy-number variants in the human genome. Nat Genet 39: S22–29, 2007. Coppinger J, Alliman S, Lamb AN, Torchia BS, Bejjani BA , Shaffer LG. Whole-genome microarray analysis in prenatal specimens identifies clinically significant chromosome alterations without increase in results of unclear significance compared to targeted microarray. Prenat Diagn 29: 1156–1166, 2009a. Coppinger J, McDonald-McGinn D, Zackai E, et al. Identification of familial and de novo microduplications of 22q11.21-q11.23 distal to the 22q11.21 microdeletion syndrome region. Hum Mol Genet 18: 1377–1383, 2009b. Córdova-Fletes C, Rademacher N, Müller I, et al. CDKL5 truncation due to a t(X;2)(p22.1;p25.3) in a girl with X-linked infantile spasm syndrome. Clin Genet 77: 92–96, 2010. Costa D, Borrell A , Margarit E, et al. Rapid fetal karyotype from cystic hygroma and pleural effusions. Prenat Diagn 15: 141–148, 1995. Costa T, Lambert M, Teshima I, Ray PN, Richer CL, Dallaire L. Monozygotic twins with 45,X/46,XY mosaicism discordant for phenotypic sex. Am J Med Genet 75: 40–44, 1998. Cotter PD, Babu A , Willner JP, Desnick RJ. Prenatal diagnosis and outcome of mosaicism for a de novo unbalanced translocation identified in amniocytes. Prenat Diagn 18: 857–861, 1998. Cotter PD, Ko E, Larabell SK , Rademaker AW, Martin RH. Segregation of a supernumerary del(15) marker chromosome in sperm. Clin Genet 58: 488–492, 2000. Cotter PD, Musci TJ. Interphase FISH with chromosome-specific protelomere probes for rapid prenatal diagnosis in a reciprocal translocation carrier. Prenat Diagn 21: 171–175, 2001. Cotter PD, Norton ME. Y chromosome heterochromatin variation detected at prenatal diagnosis. Prenat Diagn 25: 1062–1063, 2005.
532
•
REFERENCES
Cottrell C, Mendell J, Hart-Kothari M, et al. Maternal uniparental disomy of chromosome 4 in a patient with limb-girdle muscular dystrophy 2E confirmed by SNP array technology. Clin Genet in press (published online May 2011). Court Brown WM, Law P, Smith PG. Sex chromosome aneuploidy and parental age. Ann Hum Genet 33: 1–14, 1969. Courtens W, Grossman D, Van Roy N, et al. Noonanlike phenotype in monozygotic twins with a duplication-deficiency of the long arm of chromosome 18 resulting from a maternal paracentric inversion. Hum Genet 103: 497–505, 1998. Courtens W, Petersen MB, Noël JC, et al. Proximal deletion of chromosome 21 confirmed by in situ hybridization and molecular studies. Am J Med Genet 51: 260–265, 1994. Couzin DA , Watt JL, Stephen GS. Structural rearrangements in the parents of children with primary trisomy 21. J Med Genet 24: 280–282, 1987. Cowans NJ, Stamatopoulou A , Spencer K . First trimester maternal serum placental growth factor in trisomy 21 pregnancies. Prenat Diagn 30: 449–453, 2010. Cowell JK , Rutland P, Hungerford J, Jay M. Deletion of chromosome region 13q14 is transmissible and does not always predispose to retinoblastoma. Hum Genet 80: 43–45, 1988. Cox H, Stewart H, Hall L, Donnai D. Phenotypic spectrum of interstitial 7p duplication in mosaic and non-mosaic forms. Am J Med Genet 109: 306–310, 2002. Cozzi J, Conn CM, Harper J, et al. A trisomic germ cell line and precocious chromatid segregation leads to recurrent trisomy 21 conception. Hum Genet 104: 23–28, 1999. Craig-Holmes AP. C-band polymorphism in human populations. In: Hook EB, Porter IH, eds. Population Cytogenetics: Studies in Humans. New York: Academic Press, 1977: 161–177. Cram DS, Ma K , Bhasin S, et al. Y chromosome analysis of infertile men and their sons conceived through intracytoplasmic sperm injection: vertical transmission of deletions and rarity of de novo deletions. Fertil Steril 74: 909–915, 2000. Crawford DC, Acuña JM, Sherman SL. FMR1 and the fragile X syndrome: human genome epidemiology review. Genet Med 3: 359–371, 2001. Creasy MR . Complex chromosomal rearrangements. Am J Med Genet 32: 560, 1989. Crolla JA . FISH and molecular studies of autosomal supernumerary marker chromosomes excluding
those derived from chromosome 15: II. Review of the literature. Am J Med Genet 75: 367–381, 1998. Crolla JA , Howard P, Mitchell C, Long FL, Dennis NR . A molecular and FISH approach to determining karyotype and phenotype correlations in six patients with supernumerary marker(22) chromosomes. Am J Med Genet 72: 440–447, 1997. Crolla JA , Long F, Rivera H, Dennis NR . FISH and molecular study of autosomal supernumerary marker chromosomes excluding those derived from chromosomes 15 and 22: I. Results of 26 new cases. Am J Med Genet 75: 355–366, 1998. Crolla JA , van Heyningen V. Frequent chromosome aberrations revealed by molecular cytogenetic studies in patients with aniridia. Am J Hum Genet 71: 1138–1149, 2002. Crosnier C, Driancourt C, Raynaud N, et al. Mutations in JAGGED1 gene are predominantly sporadic in Alagille syndrome. Gastroenterology 116: 1141–1148, 1999. Cross I, Delhanty J, Chapman P, et al. An intrachromosomal insertion causing 5q22 deletion and familial adenomatous polyposis coli in two generations. J Med Genet 29: 175–179, 1992. Cross J, Peters G, Wu Z, Brohede J, Hannan GN. Resolution of trisomic mosaicism in prenatal diagnosis: estimated performance of a 50K SNP microarray. Prenat Diagn 27: 1197–1204, 2007. Crusi A , Engel E. Prenatal diagnosis of 3 cases of ring G chromosomes: one 21 and two 22, one of which was de novo. Ann Génét 29: 253–260, 1986. Csaba A , Bush MC, Saphier C. How painful are amniocentesis and chorionic villus sampling? Prenat Diagn 26: 35–38, 2006. Cubells JF, DeOreo EH, Harvey PD, et al. 2011. Pharmaco-genetically guided treatment of recurrent rage outbursts in an adult male with 15q13.3 deletion syndrome. Am J Med Genet 155A : 805–810, 2011. Cuckle H, Nanchahal K , Wald N. Birth prevalence of Down’s syndrome in England and Wales. Prenat Diagn 11: 29–34, 1991. Cui YX , Xia XY, Pan LJ, et al. Gonosomal mosaicism from deleted Y chromosomal nondisjunction. J Androl 28: 377–380, 2007. Cunniff C, Jones KL, Benirschke K . Ovarian dysgenesis in individuals with chromosomal abnormalities. Hum Genet 86: 552–556, 1991. Cupisti S, Conn CM, Fragouli E, et al. Sequential FISH analysis of oocytes and polar bodies reveals aneuploidy mechanisms. Prenat Diagn 23: 663–668, 2003. Curry CJ, Stevenson RE, Aughton D, et al. Evaluation of mental retardation: recommendations of a
Consensus Conference: American College of Medical Genetics. Am J Med Genet 72: 468–477, 1997. Curtis L, Antonelli E, Vial Y, et al. Prenatal diagnostic indicators of paternal uniparental disomy 14. Prenat Diagn 26: 662–666, 2006. Cutenese C, Mullett M, Hummel M, Wenger SL. Ring chromosome 1 in a newborn. Clin Dysmorphol 9: 131–133, 2000. Czakó M, Riegel M, Morava E, Schinzel A , Kosztolányi G. Patient with rheumatoid arthritis and MCA/MR syndrome due to unbalanced der(18) transmission of a paternal translocation t(18;20)(p11.1;p11.1). Am J Med Genet 108: 226–228, 2002. Dahoun S. A second case of (de novo) paracentric inversion of the short arm of the X chromosome. Ann Génét 33: 52–55, 1990. Dalby S. GIG response to the UK Clinical Genetics Society report “The genetic testing of children.” J Med Genet 32: 490–491, 1995. Dale B, Wilding M, De Matteo L, Zullo F. Absence of sperm sex chromosome aneuploidies in an X0/ XYY man. Fertil Steril 78: 634–636, 2002. Dallapiccola B, Bernardini L, Novelli A , Mingarelli R . Expanding the phenotype of duplication of the Rubinstein-Taybi region on 16p13.3. Am J Med Genet 149A : 2867–2870, 2009. Dallapiccola B, De Filippis V, Notarangelo A , Perla G, Zelante L. Ring chromosome 21 in healthy persons: different consequences in females and in males. Hum Genet 73: 218–220, 1986. Dallapiccola B, Mastroiacovo P, Gandini E. Centric fission of chromosome no. 4 in the mother of two patients with trisomy 4p. Hum Genet 31: 121–125, 1976. Dalprà L, Giardino D, Finelli P, et al. Cytogenetic and molecular evaluation of 241 small supernumerary marker chromosomes: cooperative study of 19 Italian laboratories. Genet Med 7: 620–625, 2005. Danesino C, Pasquali F, Dellavecchia C, Maserati E, Minelli A , Seghezzi L. Constitutional trisomy 8 mosaicism: mechanism of origin, phenotype variability, and risk of malignancies. Am J Med Genet 80: 540, 1998. D’Angelo CS, Da Paz JA , Kim CA , et al. PraderWilli-like phenotype: investigation of 1p36 deletion in 41 patients with delayed psychomotor development, hypotonia, obesity and/or hyperphagia, learning disabilities and behavioral problems. Eur J Med Genet 49: 451–460, 2006. Daniel A . The size of prometaphase chromosome segments. Tables using percentages of haploid autosome length (750 band stage). Clin Genet 28: 216–224, 1985.
REFERENCES
•
533
Daniel A . Distortion of female meiotic segregation and reduced male fertility in human Robertsonian translocations: consistent with the centromere model of co-evolving centromere DNA/ centromeric histone (CENP-A). Am J Med Genet 111: 450–452, 2002. Daniel A , Athayde N, Ogle R , et al. Prospective ranking of the sonographic markers for aneuploidy: data of 2143 prenatal cytogenetic diagnoses referred for abnormalities on ultrasound. Aust NZ J Obstet Gynaecol 43: 16–26, 2003a. Daniel A , Darmanian A , Peters G, Goodwin L, Hort JR . An innocuous duplication of 11.2 Mb at 13q21 is gene poor: sub-bands of gene paucity and pervasive CNV characterize the chromosome anomalies. Am J Med Genet 143A : 2452–2459, 2007. Daniel A , Hook EB, Wulf G. Collaborative U.S.A. data on prenatal diagnosis for parental carriers of chromosome rearrangements: risks of unbalanced progeny. In: A. Daniel,ed. The Cytogenetics of Mammalian Autosomal Rearrangements. New York: AR Liss, 1988: 73–162. Daniel A , Hook EB, Wulf G. Risks of unbalanced progeny at amniocentesis to carriers of chromosome rearrangements: data from United States and Canadian laboratories. Am J Med Genet 33: 14–53, 1989. Daniel A , Malafiej P. A series of supernumerary small ring marker autosomes identified by FISH with chromosome probe arrays and literature review excluding chromosome 15. Am J Med Genet 117A : 212–222, 2003. Daniel A , Ng A , Kuah KB, Reiha S, Malafiej P. A study of early amniocentesis for prenatal cytogenetic diagnosis. Prenat Diagn 18: 21–28, 1998. Daniel A , St Heaps L, Sylvester D, Diaz S, Peters G. Two mosaic terminal inverted duplications arising post-zygotically: evidence for possible formation of neo-telomeres. Cell Chromosome 7: 1, 2008. Daniel A , Wu Z, Bennetts B, et al. Karyotype, phenotype and parental origin in 19 cases of triploidy. Prenat Diagn 21: 1034–1048, 2001. Daniel A , Wu Z, Darmanian A , Collins F, Jackson J. Three different origins for apparent triploid/ diploid mosaics. Prenat Diagn 23: 529–534, 2003b. Daniel A , Wu Z, Darmanian A , et al. Issues arising from the prenatal diagnosis of some rare trisomy mosaics—the importance of cryptic fetal mosaicism. Prenat Diagn 24: 524–536, 2004. Daphnis DD, Fragouli E, Economou K , et al. Analysis of the evolution of chromosome abnormalities in human embryos from Day 3 to 5 using CGH and FISH. Mol Hum Reprod 14: 117–125, 2008.
534
•
REFERENCES
Dar H, Tal J, Bar-el H, Halpern I, Sharf M. Paracentric inversion of Xq and ovarian dysfunction. Am J Med Genet 29: 167–170, 1988. Darnell JC, Fraser CE, Mostovetsky O, Darnell RB. Discrimination of common and unique RNA-binding activities among Fragile X mental retardation protein paralogs. Hum Mol Genet 18: 3164–3177, 2009. Dauwerse JG, Hansson KBM, Brouwers AAM, Peters DJM, Breuning MH. An XX male with the sex-determining region Y gene inserted in the long arm of chromosome 16. Fertil Steril 86: 463 e1–5, 2006. Dave BJ, Olney AH, Zaleski DH, et al. Inherited 14q duplication and 21q deletion: a rare adjacent-2 segregation in multiple family members. Am J Med Genet 149A : 2248–2253, 2009. David D, Marques B, Ferreira C, et al. Characterization of two ectrodactyly-associated translocation breakpoints separated by 2.5 Mb on chromosome 2q14.1-q14.2. Eur J Hum Genet 17: 1024–1033, 2009. Davidsson J, Collin A , Öreberg M, Gisselsson D. Array-based genotype-phenotype correlation in a case of supernumerary ring chromosome 12. Clin Genet 73: 44–49, 2008. Davies AF, Kirby TL, Docherty Z, Ogilvie CM. Characterization of terminal chromosome anomalies using multisubtelomere FISH. Am J Med Genet 120A : 483–489, 2003. De Assis KR , Ladeira MS, Bueno RC, Dos Santos BF, Dalben I, Salvadori DM. Genotoxicity of cigarette smoking in maternal and newborn lymphocytes. Mutat Res 679: 72–78, 2009. De Braekeleer M, Dao TN. Cytogenetic studies in male infertility: a review. Hum Reprod 6: 245–250, 1991. De Bruijn DRH, van Dijk AHA , Pfundt R , et al. Severe progressive autism associated with two de novo changes: a 2.6-Mb 2q31.1 deletion and a balanced t(14;21)(q21.1;p11.2) translocation with long-range epigenetic silencing of LRFN5 expression. Mol Syndromol 1: 46–57, 2010. De Carvalho AFL, da Silva Bellucco FT, Kulikowski LD, Toralles MBP, Melaragno MI. Partial 5p monosomy or trisomy in 11 patients from a family with a t(5;15)(p13.3;p12) translocation. Hum Genet 124: 387–392, 2008. De Crespigny L. What’s in a name—is the pregnant woman a mother? Is the fetus a baby? Aust NZ J Obstet Gynaecol 36: 435–436, 1996. De Crespigny L, Chervenak F, McCullough L. Mothers and babies, pregnant women and fetuses. Br J Obstet Gynaecol 106: 1235–1237, 1999.
De Crespigny L, Espie M, Holmes S. Prenatal Testing: Making Choices in Pregnancy. Ringwood, Australia: Penguin Books Australia, 1998. De Decker HP, Lawrenson JB. The 22q11.2 deletion: from diversity to a single gene theory. Genet Med 3: 2–5, 2001. De Graaff E, Willemsen R , Zhong N, et al. Instability of the CGG repeat and expression of the FMR1 protein in a male fragile X patient with a lung tumor. Am J Hum Genet 57: 609–618, 1995. De Gregori M, Ciccone R , Magini P, et al. Cryptic deletions are a common finding in “balanced” reciprocal and complex chromosome rearrangements: a study of 59 patients. J Med Genet 44: 750–762, 2007. De Grouchy J, Bonnette J, Salmon C. Deletion of the short arm of chromosome 18. Ann Génét 9: 19–26, 1966. De Grouchy J, Royer P, Salmon C, Lamy M. Partial deletion of the long arms of the chromosome 18. Pathol Biol (Paris) 12: 579–582, 1964. De Grouchy J, Turleau C. Atlas des maladies chromosomiques. 2nd ed. Paris: Expansion Scientifique Française, 1982. De Jong A , Dondorp WJ, de Die-Smulders CEM, Frints SGM, de Wert GMWR . Non-invasive prenatal testing: ethical issues explored. Eur J Hum Genet 18: 272–277, 2010. De Mas P, Daudin M, Vincent MC, et al. Increased aneuploidy in spermatozoa from testicular tumour patients after chemotherapy with cisplatin, etoposide and bleomycin. Hum Reprod 16: 1204–1208, 2001. De Palma A , Burrello N, Barone N, D’Agata R , Vicari E, Calogero AE. Patients with abnormal sperm parameters have an increased sex chromosome aneuploidy rate in peripheral leukocytes. Hum Reprod 20: 2153–2156, 2005. De Pater JM, Bröcker-Vriends AHJT, Verschuren M, Linders DAC, Hansson KBM. Misleading variant chromosome 12 in prenatal diagnosis. Prenat Diagn 26: 587–588, 2006. De Pater JM, Scheres JMJC, Christiaens GCML, Ausems MGEM. Pitfalls in prenatal diagnosis: cytogenetic analysis in amniocytes fails to detect mosaic r(12). Prenat Diagn 23: 65–67, 2003a. De Pater JM, Schuring-Blom GH, van den Bogaard R , et al. Maternal uniparental disomy for chromosome 22 in a child with generalized mosaicism for trisomy 22. Prenat Diagn 17: 81–86, 1997. De Pater JM, Smeets DFCM, Scheres JMJC. Unique mosaicism of structural chromosomal rearrangement: is chromosome 18 preferentially involved? Am J Med Genet 119A : 26–31, 2003b.
De Pater JM, Van Tintelen JP, Stigter R , Brouwers HAA , Scheres JMJC. Precarious acrocentric short arm in prenatal diagnosis: no chromosome 14 polymorphism, but trisomy 17p. Genet Couns 11: 241–247, 2000. De Perdigo A , Gabriel-Robez O, Ratomponirina C, Rumpler Y. Synaptonemal complex analysis in a human male carrier of a 4;6 translocation: heterosynapsis without previous homosynapsis. Hum Genet 86: 279–282, 1991. De Perdigo A , Gabriel-Robez O, Rumpler Y. Correlation between chromosomal breakpoint positions and synaptic behaviour in human males heterozygous for a pericentric inversion. Hum Genet 83: 274–276, 1989. De Ravel TJL, Ameye L, Ballon K , Borghgraef M, Vermeesch JR , Devriendt K . Early detection of chromosome 9q22.32q31.1 microdeletion and the nevoid basal cell carcinoma syndrome. Eur J Med Genet 52: 145–147, 2009. De Ravel TJL, Keymolen K , van Assche E, et al. Post-zygotic origin of isochromosome 12p. Prenat Diagn 24: 984–988, 2004. De Ravel TJL, Legius E, Brems H, Van Hoestenberghe R , Gillis PH, Fryns JP. Hemifacial microsomia in two patients further supporting chromosomal mosaicism as a causative factor. Clin Dysmorphol 10: 263–267, 2001. De Rycke M, Liebaers I, Van Steirteghem A . Epigenetic risks related to assisted reproductive technologies: risk analysis and epigenetic inheritance. Hum Reprod 17: 2487–2494, 2002. De Silva MG, Elliott K , Dahl HH, et al. Disruption of a novel member of a sodium/hydrogen exchanger family and DOCK3 is associated with an attention deficit hyperactivity disorder-like phenotype. J Med Genet 40: 733–740, 2003. De Smith AJ, Purmann C, Walters RG, et al. A deletion of the HBII-85 class of small nucleolar RNAs (snoRNAs) is associated with hyperphagia, obesity and hypogonadism. Hum Mol Genet 18: 3257–3265, 2009. De Smith AJ, van Haelst MM, Ellis RJ, et al. Chromosome 19p13.3 deletion in a patient with macrocephaly, obesity, mental retardation, and behavior problems. Am J Med Genet 155A : 1192–1195, 2011. De Souza E, Halliday J, Chan A , Bower C, Morris JK . Recurrence risks for trisomies 13, 18, and 21. Am J Med Genet 149A : 2716–2722, 2009. De Villena FPM, Sapienza C. Transmission ratio distortion in offspring of heterozygous female carriers of Robertsonian translocations. Hum Genet 108: 31–36, 2001.
REFERENCES
•
535
De Vos A , Staessen C, De Rycke M, et al. Impact of cleavage-stage embryo biopsy in view of PGD on human blastocyst implantation: a prospective cohort of single embryo transfers. Hum Reprod 24: 2988–2996, 2009. De Vree PJP, Simon MEH, van Dooren MF, et al. Application of molecular cytogenetic techniques to clarify apparently balanced complex chromosomal rearrangements in two patients with an abnormal phenotype: case report. Mol Cytogenet 2: 15, 2009. De Vries BBA , Pfundt R , Leisink M, et al. Diagnostic genome profiling in mental retardation. Am J Hum Genet 77: 606–616, 2005. De Vries BBA , Eussen BHJ, van Diggelen OP, et al. Submicroscopic Xpter deletion in a boy with growth and mental retardation caused by a familial t(X;14). Am J Med Genet 87: 189–194, 1999. De Vries JWA , Repping S, Oates R , Carson R , Leschot NJ, van der Veen F. Absence of deleted in azoospermia (DAZ) genes in spermatozoa of infertile men with somatic DAZ deletions. Fertil Steril 75: 476–479, 2001. Dean G, Nevin NC, Mikkelsen M, et al. Investigation of a cluster of children with Down’s syndrome born to mothers who had attended a school in Dundalk, Ireland. Occup Environ Med 57: 793–804, 2000. DeBrasi D, Genardi M, D’Agostino A , et al. Double autosomal/gonosomal mosaic aneuploidy: study of nondisjunction in two cases with trisomy of chromosome 8. Hum Genet 95: 519–525, 1995. Dee SL, Clark AT, Willatt LR , Yates JRW. A case of ring chromosome 2 with growth retardation, mild dysmorphism, and microdeletion of 2p detected using FISH. J Med Genet 38: E32, 2001. Del Porto G, Di Fusco C, Baldi M, Grammatico P, D’Alessandro E. Familial centric fission of chromosome 4. J Med Genet 21: 388–391, 1984. Delatycki M, Gardner RJM. Three cases of trisomy 13 mosaicism and a review of the literature. Clin Genet 51: 403–407, 1997. Delatycki MB, Pertile MD, Gardner RJM. Trisomy 13 mosaicism at prenatal diagnosis: dilemmas in interpretation. Prenat Diagn 18: 45–50, 1998. Delhanty J, Conn C. Preimplantation genetic diagnosis of chromosome abnormalities: specific chromosomal rearrangements and age-related aneuploidy. In: Harper JC, Delhanty JDA , Handyside AH, eds. Preimplantation Genetic Diagnosis. Chichester, England: John Wiley & Sons, 2001: 203–223. Delisle MF, Wilson RD. First trimester prenatal diagnosis: amniocentesis. Semin Perinatol 23: 414–423, 1999.
536
•
REFERENCES
Delobel B, Djlelati R , Gabriel-Robez O, et al. Y-autosome translocation and infertility: usefulness of molecular, cytogenetic and meiotic studies. Hum Genet 102: 98–102, 1998. DeLozier-Blanchet CD. Genetic counseling and prenatal diagnosis. In: Engel E, Antonarakis SE, eds. Genomic Imprinting and Uniparental Disomy in Medicine: Clinical and Molecular Aspects. New York: Wiley-Liss, 2002: 227–242. DeLozier-Blanchet CD, Roeder E, Denis-Arrue R , et al. Trisomy 12 mosaicism confirmed in multiple organs from a liveborn child. Am J Med Genet 95: 444–449, 2000. Dempsey MA , Schwartz S, Waggoner DJ. Mosaicism del(22)(q11.2q11.2)/dup(22)(q11.2q11.2) in a patient with features of 22q11.2 deletion syndrome. Am J Med Genet 143A : 1082–1086, 2007. Denayer E, Brems H, de Cock P, et al. Pathogenesis of vestibular schwannoma in ring chromosome 22. BMC Med Genet 10: 97, 2009. Deng HX , Abe K , Kondo I, et al. Parental origin and mechanism of formation of polysomy X: an XXXXX case and four XXXXY cases determined with RFLPs. Hum Genet 86: 541–544, 1991. Denschlag D, Tempfer C, Kunze M, Wolff G, Keck C. Assisted reproductive techniques in patients with Klinefelter syndrome: a critical review. Fertil Steril 82: 775–779, 2004. DeScipio C, Morrissette JD, Conlin LK , et al. Two siblings with alternate unbalanced recombinants derived from a large cryptic maternal pericentric inversion of chromosome 20. Am J Med Genet 152A : 373–382, 2010a. DeScipio C, Morrissette JD, Conlin LK , et al. Update on “Two siblings with alternate unbalanced recombinants derived from a large cryptic maternal pericentric inversion of chromosome 20.” Am J Med Genet 152A :1599, 2010b. DesGroseilliers M, Beaulieu Bergeron M, Brochu P, Lemyre E, Lemieux N. Phenotypic variability in isodicentric Y patients: study of nine cases. Clin Genet 70: 145–150, 2006. Desmyttere S, De Schepper J, Nekkebroeck J, et al. Two-year auxological and medical outcome of singletons born after embryo biopsy applied in preimplantation genetic diagnosis or preimplantation genetic screening. Hum Reprod 24: 470–476, 2009. Destree A , Fourneau C, Dugauquier C, Rombout S, Sartenaer D, Gillerot Y. Prenatal diagnosis of trisomy 6 mosaicism. Prenat Diagn 25: 354–357, 2005. Deveault C, Qian JH, Chebaro W, et al. NLRP7 mutations in women with diploid androgenetic
and triploid moles: a proposed mechanism for mole formation. Hum Mol Genet 18: 888–897, 2009. Devi A , Benn PA . X-chromosome abnormalities in women with premature ovarian failure. J Reprod Med 44: 321–324, 1999. Devillard F, Metzler-Guillemain C, Pelletier R , et al. Polyploidy in large-headed sperm: FISH study of three cases. Hum Reprod 17: 1292–1298, 2002. Devine DH, Whitman-Elia G, Best RG, Edwards JG. Paternal paracentric inversion of chromosome 2: a possible association with recurrent pregnancy loss and infertility. J Assist Reprod Genet 17: 293–296, 2000. Devriendt K , Van Hoestenberghe R , Van Hole C, et al. Submicroscopic deletion in chromosome 22q11 in trizygous triplet siblings and their father. Clinical variability of 22q11 deletion. Clin Genet 51: 246–249, 1997. Devroey P, Wisanto A , Camus M, et al. Oocyte donation in patients without ovarian function. Hum Reprod 3: 699–704, 1988. Dewhurst J. Fertility in 47,XXX and 45,X patients. J Med Genet 15: 132–135, 1978. Dhallan R , Guo X , Emche S, et al. A non-invasive test for prenatal diagnosis based on fetal DNA present in maternal blood: a preliminary study. Lancet 369: 474–481, 2007. Dhandha S, Hogge WA , Surti U, McPherson E. Three cases of tetrasomy 9p. Am J Med Genet 113: 375–380, 2002. D’Hulst C, Kooy RF. Fragile X syndrome: from molecular genetics to therapy. J Med Genet 46: 577–584, 2009. Dibbens LM, Mullen S, Helbig I, et al. Familial and sporadic 15q13.3 microdeletions in idiopathic generalized epilepsy: precedent for disorders with complex inheritance. Hum Mol Genet 18: 3626–3631, 2009. Diego-Alvarez D, Ramos-Corrales C, Garcia-Hoyos M, et al. Double trisomy in spontaneous miscarriages: cytogenetic and molecular approach. Hum Reprod 21: 958–966, 2006. Diego-Alvarez D, Rodriguez de Alba M, CarderoMerlo R , et al. MLPA as a screening method of aneuploidy and unbalanced chromosomal rearrangements in spontaneous miscarriages. Prenat Diagn 27: 765–771, 2007. Digilio MC, Bernardini L, Mingarelli R , et al. 3q29 Microdeletion: a mental retardation disorder unassociated with a recognizable phenotype in two mother-daughter pairs. Am J Med Genet 149A : 1777–1781, 2009. Dirnfeld M, Bider D, Abramovicia H, Calderon I, Blumenfeld Z. Subsequent successful pregnancy
and delivery after intracytoplasmic sperm injection in a patient with XY gonadal dysgenesisms. Eur J Obstet Gynecol Reprod Biol 88: 101–102, 2000. DiPietro JA , Cristofalo EA , Voegtline KM, Crino J. Isolated prenatal choroid plexus cysts do not affect child development. Prenat Diagn 31: 745–749, 2011. Disteche CM. Escape from X inactivation in human and mouse. Trends Genet 11: 17–22, 1995. Dobkin C, Radu G, Ding XH, Brown WT, Nolin SL. Fragile X prenatal analyses show full mutation females at high risk for mosaic Turner syndrome: fragile X leads to chromosome loss. Am J Med Genet 149A : 2152–2157, 2009. Dobyns WB, Mirzaa G, Christian SL, et al. Consistent chromosome abnormalities identify novel polymicrogyria loci in 1p36.3, 2p16.1-p23.1, 4q21.21-q22.1, 6q26-q27, and 21q2. Am J Med Genet 146A : 1637–1654, 2008. Doheny KF, Rasmussen SA , Rutberg J, et al. Segregation of a familial balanced (12;10) insertion resulting in Dup(10)(q21.2q22.1) and Del(10)(q21.2q22.1) in first cousins. Am J Med Genet 69: 188–193, 1997. Dohle GR , Halley DJJ, Van Hemel JO, et al. Genetic risk factors in infertile men with severe oligozoospermia and azoospermia. Hum Reprod 17: 13–16, 2002. Dolan M, Mendelsohn NJ, Pierpont ME, Schimmenti LA , Berry SA , Hirsch B. A novel microdeletion/microduplication syndrome of 19p13.13. Genet Med 12: 503–511, 2010. Dominguez MG, Rivera H, Vasquez AI, HernandezZaragoza G, Rivas F. Interchange trisomy 21 by t(1;21)(p22;q22)mat. Genet Couns 12: 363–367, 2001. Donaghue C, Mann K , Docherty Z, Mackie Ogilvie C. Detection of mosaicism for primary trisomies in prenatal samples by QF-PCR and karyotype analysis. Prenat Diagn 25: 65–72, 2005. Donnai D, Karmiloff-Smith A . Williams syndrome: from genotype through to the cognitive phenotype. Am J Med Genet 97: 164–171, 2000. Donnelly SL, Wolpert CM, Menold MM, et al. Female with autistic disorder and monosomy X (Turner syndrome): parent-of-origin effect of the X chromosome. Am J Med Genet 96: 312–316, 2000. Donoso P, Staessen C, Fauser BC, Devroey P. Current value of preimplantation genetic aneuploidy screening in IVF. Hum Reprod Update 13: 15–25, 2007. Draaken M, Reutter H, Schramm C, et al. Microduplications at 22q11.21 are associated with non-syndromic classic bladder exstrophy. Eur J Med Genet 53: 55–60, 2010.
REFERENCES
•
537
Drugan A , Greb A , Johnson MP, et al. Determinants of parental decisions to abort for chromosome abnormalities. Prenat Diagn 10: 483–490, 1990. Duba HC, Weirich HG, Weirich-Schwaiger H, et al. Chromosomal instability in a woman with infertility and two unaffected brothers: a new familial chromosomal breakage syndrome? Hum Genet 100: 431–440, 1997. Duckett DP, Roberts SH. Adjacent 2 meiotic disjunction. report of a case resulting from a familial 13q;15q balanced reciprocal translocation and review of the literature. Hum Genet 58: 377–386, 1981. Dufke A , Eggermann K , Balg S, Stengel-Rutkowski S, Enders H, Kaiser P. A second case of inv(4)pat with both recombinants in the offspring: rec dup(4q) in a girl with Wolf-Hirschhorn syndrome and rec dup(4p). Cytogenet Cell Genet 91: 85–89, 2000. Dufke A, Mayrhofer H, Enders H, Kaiser P, Leipoldt M. Unusual chromosomal mosaicism as a cause of mental retardation and congenital malformations in a familial reciprocal translocation carrier, t(17;22)(q24.2;q11.23). Cytogenet Cell Genet 93: 168–170, 2001. Duker NJ. Chromosome breakage syndromes and cancer. Am J Med Genet 115: 125–129, 2002. Dundar M, Caglayan AO, Saatci C, Batukan C, Basbug M, Ozkul Y. Can the classical euchromatic variants of 9q12/qh+ cause recurrent abortions? Genet Couns 19: 281–286, 2008. Dunn TM, Grunfeld L, Kardon NB. Trisomy 1 in a clinically recognized IVF pregnancy. Am J Med Genet 99: 152–153, 2001. Dupont C, Bavister BD, Armant DR , Brenner CA . Rhesus macaque embryos derived from MI oocytes maturing after retrieval display high rates of chromosomal anomalies. Hum Reprod 24: 929–935, 2009. Dupont C, Delahaye A , Burglen L, et al. First cryptic balanced reciprocal translocation mosaicism and familial transmission. Am J Med Genet 146A : 2971–2974, 2008. Dupont JM, Cuisset L, Cartigny M, et al. Familial reciprocal translocation t(7;16) associated with maternal uniparental disomy 7 in a Silver-Russell patient. Am J Med Genet 111: 405–408, 2002. Durante M, Snigiryova G, Akaeva E, et al. Chromosome aberration dosimetry in cosmonauts after single or multiple space flights. Cytogenet Genome Res 103: 40–46, 2003. Durban M, Benet J, Boada M, et al. PGD in female carriers of balanced Robertsonian and reciprocal translocations by first polar body analysis. Hum Reprod Update 7: 591–602, 2001.
538
•
REFERENCES
Du Sart D, Kalitsis P, Schmidt M. Noninactivation of a portion of Xq28 in a balanced X-autosome translocation. Am J Med Genet 42: 156–160, 1992. Earle E, Voullaire LE, Hills L, Slater H, Choo KHA . Absence of satellite III DNA in the centromere and the proximal long-arm region of human chromosome 14: analysis of a 14p- variant. Cytogenet Cell Genet 61: 78–80, 1992. Eckel H, Wimmer R , Volleth M, Jakubiczka S, Muschke P, Wieacker P. Intrachromosomal triplication 12p11.22-p12.3 and gonadal mosaicism of partial tetrasomy 12p. Am J Med Genet 140: 1219–1222, 2006. Edwards JH. Familiarity, recessivity and germline mosaicism. Ann Hum Genet 53: 33–47, 1989. Edwards MJ, Park JP, Wurster-Hill DH, Graham JM, Jr. Mixoploidy in humans: two surviving cases of diploid-tetraploid mixoploidy and comparison with diploid-triploid mixoploidy. Am J Med Genet 52: 324–330, 1994. Egan JFX , Smith K , Timms D, Bolnick JM, Campbell WA , Benn PA. Demographic differences in Down syndrome livebirths in the US from 1989 to 2006. Prenat Diagn 31: 389–394, 2011. Eggermann T, Begemann M, Binder G, Spengler S. Silver-Russell syndrome: genetic basis and molecular genetic testing. Orphanet J Rare Dis 5: 19, 2010a. Eggermann T, Curtis M, Zerres K , Hughes HE. Maternal uniparental disomy 16 and genetic counseling: new case and survey of published cases. Genet Couns 15: 183–190, 2004. Eggermann T, Engels H, Heidrich-Kaul C, Moderau I, Schwanitz G. Molecular investigation of the parental origin of a de novo unbalanced translocation 13/18. Hum Genet 99: 521–522, 1997. Eggermann T, Gonzalez D, Spengler S, ArslanKirchner M, Binder G, Schonherr N. Broad clinical spectrum in Silver-Russell syndrome and consequences for genetic testing in growth retardation. Pediatrics 123: e929–931, 2009. Eggermann K , Mau UA , Bujdosó G, et al. Supernumerary marker chromosomes derived from chromosome 15: analysis of 32 new cases. Clin Genet 62: 89–93, 2002. Eggermann T, Mergenthaler S, Eggermann K , et al. Identification of interstitial maternal uniparental disomy (UPD) (14) and complete maternal UPD(20) in a cohort of growth retarded patients. J Med Genet 38: 86–89, 2001. Eggermann T, Rossier E, Theurer-Mainka U, et al. New case of mosaic tetrasomy 9p with additional neurometabolic findings. Am J Med Genet 75: 530–533, 1998.
Eggermann T, Schönherr N, Spengler S, et al. Identification of a 21q22 duplication in a Silver-Russell syndrome patient further narrows down the Down syndrome critical region. Am J Med Genet 152A : 356–359, 2010b. Eggermann T, Spengler S, Bachmann N, et al. Chromosome 11p15 duplication in Silver-Russell syndrome due to a maternally inherited translocation t(11;15). Am J Med Genet 152A : 1484–1487, 2010c. Egozcue S, Blanco J, Vendrell JM, et al. Human male infertility: chromosome anomalies, meiotic disorders, abnormal spermatozoa and recurrent abortion. Hum Reprod Update 6: 93–105, 2000. Ehrenheim C, Hauswirth C, Fitschen J, Martin E, Oetting G, Hundeshagen H. Genetic risk after high dose radioiodine therapy with regard to gonadal dose. Nuklearmedizin 36: 157–166, 1997. Ehrlich M, Tsien F, Herrera D, Blackman V, Roggenbuck J, Tuck-Muller CM. High frequencies of ICF syndrome-like pericentromeric heterochromatin decondensation and breakage in chromosome 1 in a chorionic villus sample. J Med Genet 38: 882–884, 2001. Ekelund CK , Jørgensen FS, Petersen OB, Sundberg K , Tabor A . Impact of a new national screening policy for Down’s syndrome in Denmark: population based cohort study. Br Med J 337: a2547, 2008. El-Fouly MH, Higgins JV, Kapur S, Sankey BJ, Matisoff DN, Costa-Fox M. DiGeorge anomaly in an infant with deletion of chromosome 22 and dup(9p) due to adjacent type II disjunction. Am J Med Genet 38: 569–573, 1991. Elghezal H, Hidar S, Braham R , Denguezli W, Ajina M, Saad A . Chromosome abnormalities in one thousand infertile males with nonobstructive sperm disorders. Fertil Steril 86: 1792–1795, 2006. Eliez S, Morris MA , Dahoun-Hadorn S, et al. Familial translocation t(Y;15)(q12;p11) and de novo deletion of the Prader-Willi syndrome (PWS) critical region on 15q11-q13. Am J Med Genet 70: 222–228, 1997. Elkins TE, Gafford LS, Wilks CS, Muram D, Golden G. A model clinic approach to the reproductive health concerns of the mentally handicapped. Obstet Gynecol 68: 185–188, 1986a. Elkins TE, Stovall TG, Wilroy S, Dacus JV. Attitudes of mothers of children with Down syndrome concerning amniocentesis, abortion, and prenatal genetic counseling techniques. Obstet Gynecol 68: 181–184, 1986b. Elliott M, Bayly R , Cole T, Temple IK , Maher ER . Clinical features and natural history of BeckwithWiedemann syndrome: presentation of 74 new cases. Clin Genet 46: 168–174, 1994.
Ellis NA , Ciocci S, German J. Back mutation can produce phenotype reversion in Bloom syndrome somatic cells. Hum Genet 108: 167–173, 2001. Ellison JW, Tekin M, Sikes KS, et al. Molecular characterization of a ring X chromosome in a male with short stature. Hum Genet 110: 322–326, 2002. Emery AEH. Methodology in Medical Genetics: An Introduction to Statistical Methods. Edinburgh, Scotland: Churchill Livingstone, 1986. Emiliani S, Gonzalez-Merino E, Van den Bergh M, Abramowicz M, Englert Y. Higher degree of chromosome mosaicism in preimplantation embryos from carriers of robertsonian translocation t(13;14) in comparison with embryos from karyotypically normal IVF patients. J Assist Reprod Genet 20: 95–100, 2003. Engel E. A new genetic concept: uniparental disomy and its potential effect, isodisomy. Am J Med Genet 6: 137–143, 1980. Engel E, Antonarakis SE. Genomic Imprinting and Uniparental Disomy: Clinical and Molecular Aspects. New York: Wiley-Liss, 2002. Engel E, Crippa L, Engel-de Montmollin M, Tran TN, Muhlethaler M. Implications for genetic counselling in regard to sex chromosome aneuploidies diagnosed by amniocentesis. Ann Génét 24: 107–109, 1981. Engelen JJM, Arens YHJM, Gondrie ETCM, Alofs MGP, Loneus WH, Hamers AJH. Intrachromosomal insertion translocation resulting in duplication of chromosome band Yq11.2 in two fertile brothers. Am J Med Genet 118A : 287–289, 2003. Engelen JJM, Marcelis C, Herbergs J, et al. Mosaic telomeric (2;14) association in a child with motor delay. Am J Med Genet 92: 318–321, 2000. Engels H, Eggermann T, Caliebe A , et al. Genetic counseling in Robertsonian translocations der(13;14): frequencies of reproductive outcomes and infertility in 101 pedigrees. Am J Med Genet 146A : 2611–2616, 2008. Engenheiro E, Moller RS, Pinto M, et al. MowatWilson syndrome: an underdiagnosed syndrome? Clin Genet 73: 579–584, 2008. Ensenauer RE, Adeyinka A , Flynn HC, et al. Microduplication 22q11.2, an emerging syndrome: clinical, cytogenetic, and molecular analysis of thirteen patients. Am J Hum Genet 73: 1027–1040, 2003. Epstein CJ. The Phenotypic Mapping of Down Syndrome and Other Aneuploid Conditions. New York: Wiley-Liss, 1993. Epstein CJ. 2001 William Allan Award Address. From Down syndrome to the “human” in “human genetics.” Am J Hum Genet 70: 300–313, 2002.
REFERENCES
•
539
Ergul E, Liehr T, Mrasek K , Sazci A . A de novo complex chromosome rearrangement involving three chromosomes (2, 13, and 18) in an oligospermic male. Fertil Steril 92: 391 e12, 2009. Erickson RP, Hudgins L, Stone JF, Schmidt S, Wilke C, Glover TW. A “balanced” Y;16 translocation associated with Turner-like neonatal lymphedema suggests the location of a potential anti-Turner gene on the Y chromosome. Cytogenet Cell Genet 71: 163–167, 1995. Escribá MJ, Martín J, Rubio C, et al. Heteroparental blastocyst production from microsurgically corrected tripronucleated human embryos. Fertil Steril 86: 1601–1607, 2006. Escudero T, Abdelhadi I, Sandalinas M, Munne S. Predictive value of sperm fluorescence in situ hybridization analysis on the outcome of preimplantation genetic diagnosis for translocations. Fertil Steril 79 Suppl 3: 1528–1534, 2003. Escudero T, Estop A , Fischer J, Munné S. Preimplantation genetic diagnosis for complex chromosome rearrangements. Am J Med Genet 146A : 1662–1669, 2008. Escudero T, Lee M, Sandalinas M, Munné S. Female gamete segregation in two carriers of translocations involving 2q and 14q. Prenat Diagn 20: 235–237, 2000. Escudero T, Lee M, Stevens J, Sandalinas M, Munné S. Preimplantation genetic diagnosis of pericentric inversions. Prenat Diagn 21: 760–766, 2001. Eskenazi B, Wyrobek AJ, Kidd SA , et al. Sperm aneuploidy in fathers of children with paternally and maternally inherited Klinefelter syndrome. Hum Reprod 17: 576–583, 2002. Esmer C, Sánchez S, Ramos S, Molina B, Frias S, Carnevale A . DEB test for Fanconi anemia detection in patients with atypical phenotypes. Am J Med Genet 124A : 35–39, 2004. Estop AM, Cieply KM, Munné S, Feingold E. Multicolor fluorescence in situ hybridization analysis of the spermatozoa of a male heterozygous for a reciprocal translocation t(11;22)(q23;q11). Hum Genet 104: 412–417, 1999. Estop AM, Cieply K , Munne S, Surti U, Wakim A , Feingold E. Is there an interchromosomal effect in reciprocal translocation carriers? Sperm FISH studies. Hum Genet 106: 517–524, 2000. European Collaborative Research on Mosaicism in CVS (EUCROMIC). Trisomy 15 CPM: probable origins, pregnancy outcome and risk of fetal UPD. Prenat Diagn 19: 29–35, 1999. European Polycystic Kidney Disease Consortium. The polycystic kidney disease 1 gene encodes a 14 kb
540
•
REFERENCES
transcript and lies within a duplicated region on chromosome 16. Cell 77: 881–894, 1994. Eussen BHJ, Bartalini G, Bakker L, et al. An unbalanced submicroscopic translocation t(8;16) (q24.3;p13.3)pat associated with tuberous sclerosis complex, adult polycystic kidney disease, and hypomelanosis of Ito. J Med Genet 37: 287–291, 2000. Evans JA , MacDonald K , Hamerton JL. Sex chromosome anomalies: prenatal diagnosis and the need for continued prospective studies. In: Evans JA, Hamerton JL, Robinson A, eds. Children and Young Adults with Sex Chromosome Aneuploidy: Follow-up, Clinical, and Molecular Studies. Birth Defects: Orig Art Series 26(4): 273–281, 1990. Evans MI, Farrell SA , Greb A , Ray P, Johnson MP, Hoffman EP. In utero fetal muscle biopsy for the diagnosis of Duchenne muscular dystrophy in a female fetus “suddenly at risk .” Am J Med Genet 46: 309–312, 1993. Evans MI, White BJ, Kent SG, Levine MA , Levin SW, Larsen JW, Jr. Balanced rearrangement of chromosomes 2, 5, and 13 in a family with duplication 5q and fetal loss. Am J Med Genet 19: 783–790, 1984. Everman DB, Morgan CT, Lyle R , et al. Frequency of genomic rearrangements involving the SHFM3 locus at chromosome 10q24 in syndromic and non-syndromic split-hand/foot malformation. Am J Med Genet 140: 1375–1383, 2006. Faas BHW, van der Burgt I, Kooper AJA , et al. Identification of clinically significant, submicroscopic chromosome alterations and UPD in fetuses with ultrasound anomalies using genome-wide 250k SNP array analysis. J Med Genet 47: 586–594, 2010. Faivre L, Gosset P, Cormier-Daire V, et al. Overgrowth and trisomy 15q26.1-qter including the IGF1 receptor gene: report of two families and review of the literature. Eur J Hum Genet 10: 699–706, 2002. Faivre L, Morichon-Delvallez N, Viot G, et al. Prenatal diagnosis of a satellited non-acrocentric chromosome derived from a maternal translocation (10;13)(p13;p12) and review of literature. Prenat Diagn 19: 282–286, 1999. Faivre L, Portnoï MF, Pals G, et al. Should chromosome breakage studies be performed in patients with VACTERL association? Am J Med Genet 137: 55–58, 2005. Faivre L, Radford I, Viot G, et al. Cerebellar ataxia and mental retardation in a child with an inherited satellited chromosome 4q. Ann Génét 43: 35–38, 2000a. Faivre L, Viot G, Prieur M, et al. Apparent Sotos syndrome (cerebral gigantism) in a child with
trisomy 20p11.2-p12.1 mosaicism. Am J Med Genet 91: 273–276, 2000b. Falik-Borenstein TC, Korenberg JR , Schreck RR . Confined placental chimerism: prenatal and postnatal cytogenetic and molecular analysis, and pregnancy outcome. Am J Med Genet 50: 51–56, 1994. Falik-Borenstein TC, Pribyl TM, Pulst SM, et al. Stable ring chromosome 21: molecular and clinical definition of the lesion. Am J Med Genet 42: 22–28, 1992. Familiari G, Caggiati A , Nottola SA , Ermini M, Di Benedetto MR , Motta PM. Ultrastructure of human ovarian primordial follicles after combination chemotherapy for Hodgkin’s disease. Hum Reprod 8: 2080–2087, 1993. Fan YS, Siu VM, Jung JH, Farrell SA , Côté GB. Direct duplication of 8p21.3→p23. 1: a cytogenetic anomaly associated with developmental delay without consistent clinical features. Am J Med Genet 103: 231–234, 2001. Fantes JA , Boland E, Ramsay J, et al. FISH mapping of de novo apparently balanced chromosome rearrangements identifies characteristics associated with phenotypic abnormality. Am J Hum Genet 82: 916–926, 2008. Fantes JA , Mewborn SK , Lese CM, et al. Organisation of the pericentromeric region of chromosome 15: at least four partial gene copies are amplified in patients with a proximal duplication of 15q. J Med Genet 39: 170–177, 2002. Farag TI, Krishna Murthy DS, Al-Awadi SA , et al. Robertsonian translocation t dic (14p;22p) with regular trisomy 21: a possible interchromosomal effect? Ann Génét 30: 189–192, 1987. Farah LMS, Nazareth HRdeS, Dolnikoff M, Delascio D. Balanced homologous translocation t(22q22q) in a phenotypically normal woman with repeated spontaneous abortions. Humangenetik 28: 357–360, 1975. Farah SB, Ramos CF, de Mello MP, et al. Two cases of Y; autosome translocations: A 45,X male and a clinically trisomy 18 patient. Am J Med Genet 49: 388–392, 1994. Faraut T, Mermet MA , Demongeot J, Cohen O. Cooperation of selection and meiotic mechanisms in the production of imbalances in reciprocal translocations. Cytogenet Cell Genet 88: 15–21, 2000. Faravelli F, Murdolo M, Marangi G, Bricarelli FD, Di Rocco M, Zollino M. Mother to son amplification of a small subtelomeric deletion: a new mechanism of familial recurrence in microdeletion syndromes. Am J Med Genet 143A : 1169–1173, 2007.
Farlie P, Reid C, Wilcox S, Peeters J, Reed G, Newgreen D. Ypel 1: a novel nuclear protein that induces an epithelial-like morphology in fibroblasts. Genes Cells 6: 619–629, 2001. Farrell SA , Chow G. Intrachromosomal insertion of chromosome 7. Clin Genet 41: 299–302, 1992. Farrell SA , Fan YS. Balanced nonacrocentric wholearm reciprocal translocations: a de novo case and literature review. Am J Med Genet 55: 423–426, 1995. Farrell SA , Summers AM, Gardner HA , Uchida IA . Balanced complex chromosome rearrangement ascertained through prenatal diagnosis. Am J Med Genet 52: 360–361, 1994. Farrell SA , Winsor EJT, Markovic VD. Moving satellites and unstable chromosome translocations: clinical and cytogenetic implications. Am J Med Genet 46: 715–720, 1993. Federman DD. Mapping the X-chromosome: mapping its p’s and q’s. N Engl J Med 317: 161–162, 1987. Feenstra I, Fang J, Koolen DA , et al. European Cytogeneticists Association Register of Unbalanced Chromosome Aberrations (ECARUCA); an online database for rare chromosome abnormalities. Eur J Med Genet 49: 279–291, 2006. Fejgin MD, Arbel-DeRowe Y, Shul N, Amiel A . A false-positive diagnosis of Turner syndrome by amniocentesis. Prenat Diagn 17: 88–89, 1997. Feldman B, Ebrahim SAD, Hazan SL, et al. Routine prenatal diagnosis of aneuploidy by FISH studies in high-risk pregnancies. Am J Med Genet 90: 233–238, 2000. Feldman B, Kramer RL, Ebrahim SAD, Wolff DJ, Evans MI. Prenatal evaluation of a de novo X;9 translocation. Am J Med Genet 85: 476–478, 1999. Feldman GL, Weiss L, Phelan MC, Schroer RJ, Van Dyke DL. Inverted duplication of 8p: ten new patients and review of the literature. Am J Med Genet 47: 482–486, 1993. Fellermann K , Stange DE, Schaeffeler E, et al. A chromosome 8 gene-cluster polymorphism with low human beta-defensin 2 gene copy number predisposes to Crohn disease of the colon. Am J Hum Genet 79: 439–448, 2006. Feng Y, Zhang F, Lokey LK , et al. Translational suppression by trinucleotide repeat expansion at FMR1. Science 268: 731–734, 1995. Fénichel P, Letur H. Procréation et syndrome de Turner: quelles recommandations avant, pendant et après la grossesse? Gynécol Obstét Fertil 36: 891–897, 2008. Ferfouri F, Clement P, Molina Gomes D, et al. Is classic pericentric inversion of chromosome 2
REFERENCES
•
541
inv(2)(p11q13) associated with an increased risk of unbalanced chromosomes? Fertil Steril 92: 1497 e1–4, 2009. Ferguson-Smith MA . Prenatal chromosome analysis and its impact on the birth incidence of chromosome disorders. Br Med Bull 39: 355–364, 1983. Ferguson-Smith MA , Cooke A , Affara NA , Boyd E, Tolmie JL. Genotype-phenotype correlations in XX males and their bearing on current theories of sex determination. Hum Genet 84: 198–202, 1990. Ferhi K , Avakian R , Griveau JF, Guille F. Age as only predictive factor for successful sperm recovery in patients with Klinefelter’s syndrome. Andrologia 41: 84–87, 2009. Fernandez BA , Roberts W, Chung B, et al. Phenotypic spectrum associated with de novo and inherited deletions and duplications at 16p11.2 in individuals ascertained for diagnosis of autism spectrum disorder. J Med Genet 47: 195–203, 2010. Fernandez-Carvajal I, Lopez Posadas B, Pan R , Raske C, Hagerman PJ, Tassone F. Expansion of an FMR1 grey-zone allele to a full mutation in two generations. J Mol Diagn 11: 306–310, 2009. Fernandez-Carvajal I, Walichiewicz P, Xiaosen X , Pan R , Hagerman PJ, Tassone F. Screening for expanded alleles of the FMR1 gene in blood spots from newborn males in a Spanish population. J Mol Diagn 11: 324–329, 2009b. Fernández-Novoa MC, Vargas MT, Vizmanos JL, et al. Prader-Willi syndrome large deletion on two brothers. Is this the exception that confirm the rule? Rev Neurol 32: 935–938, 2001. Ferraretti AP, Magli MC, Kopcow L, Gianaroli L. Prognostic role of preimplantation genetic diagnosis for aneuploidy in assisted reproductive technology outcome. Hum Reprod 19: 694–699, 2004. Ferrero GB, Howald C, Micale L, et al. An atypical 7q11.23 deletion in a normal IQ Williams-Beuren syndrome patient. Eur J Hum Genet 18: 33–38, 2010. Ferrier RA , Lowry RB, Lemire EG, Stoeber GP, Howard J, Parboosingh JS. Father-to-son transmission of an X-linked gene: a case of paternal sex chromosome heterodisomy. Am J Med Genet 149A : 2871–2873, 2009. Fewtrell MS, Tam PKH, Thomson AH, et al. Hirschsprung’s disease associated with a deletion of chromosome 10 (q11.2q21.2): a further link with the neurocristopathies? J Med Genet 31: 325–327, 1994. Field LL, Tobias R , Robinson WP, Paisey R , Bain S. Maternal uniparental disomy of chromosome 1
542
•
REFERENCES
with no apparent phenotypic effects. Am J Hum Genet 63: 1216–1220, 1998. Filges I, Kang A, Klug V, et al. aCGH on chorionic villi mirrors the complexity of fetoplacental mosaicism in prenatal diagnosis. Prenat Diagn 31 :473–478, 2011. Filges I, Röthlisberger B, Wenzel F, Heinimann K , Huber AR , Miny P. Mosaic ring chromosome 8: clinical and array-CGH findings in partial trisomy 8. Am J Med Genet 146A : 2837–2841, 2008. Filho AB, Souza J, Faucz FR , et al. Somatic/gonadal mosaicism in a syndromic form of ectrodactyly, including eye abnormalities, documented through array-based comparative genomic hybridization. Am J Med Genet 155A : 1152–1156, 2011. Fineman RM, Issa B, Weinblatt V. Prenatal diagnosis of a large heteromorphic region in a chromosome 5: implications for genetic counseling. Am J Med Genet 32: 498–499, 1989. Fiorentino F, Spizzichino L, Bono S, et al. PGD for reciprocal and Robertsonian translocations using array comparative genomic hybridization. Hum Reprod 26: 1925–1935, 2011. Firth H. Chorion villus sampling and limb deficiency—cause or coincidence? Prenat Diagn 17: 1313–1330, 1997. Fischbach BV, Trout KL, Lewis J, Luis CA , Sika M. WAGR syndrome: a clinical review of 54 cases. Pediatrics 116: 984–988, 2005. Fisher RA , Khatoon R , Paradinas FJ, Roberts AP, Newlands ES. Repetitive complete hydatidiform mole can be biparental in origin and either male or female. Hum Reprod 15: 594–598, 2000. Fitzgerald PH, Archer SA , Morris CM. Evidence for the repeated primary non-disjunction of chromosome 21 as a result of premature centromere division (PCD). Hum Genet 72: 58–62, 1986. Fitzgerald PH, Donald RA , McCormick P. Reduced fertility in women with X chromosome abnormality. Clin Genet 25: 301–309, 1984. Flejter WL, Finlinson D, Root S, Nguyen W, Brothman AR , Viskochil D. Familial ring (19) chromosome mosaicism: case report and review. Am J Med Genet 66: 276–280, 1996. Flipsen-ten Berg K , van Hasselt PM, Eleveld MJ, et al. Unmasking of a hemizygous WFS1 gene mutation by a chromosome 4p deletion of 8.3 Mb in a patient with Wolf-Hirschhorn syndrome. Eur J Hum Genet 15: 1132–1138, 2007. Flori E, Doray B, Rudolf G, et al. Failure of prenatal diagnosis of diploid-triploid mosaicism after amniocentesis. Clin Genet 63: 328–331, 2003. Forabosco A , Percesepe A , Santucci S. Incidence of non-age-dependent chromosomal abnormalities: a
population-based study on 88965 amniocenteses. Eur J Hum Genet 17: 897–903, 2009. Ford CE, Jones KW, Miller OJ, et al. The chromosomes in a patient showing both mongolism and the Klinefelter syndrome. Lancet 1: 709–710, 1959. Ford NM. When did I begin? Cambridge, England: Cambridge University Press, 1988. Foresta C, Ferlin A , Gianaroli L, Dallapiccola B. Guidelines for the appropriate use of genetic tests in infertile couples. Eur J Hum Genet 10: 303–312, 2002. Foresta C, Moro E, Ferlin A . Y chromosome microdeletions and alterations of spermatogenesis. Endocr Rev 22: 226–239, 2001. Forrest LE, Delatycki MB, Skene L, Aitken M. Communicating genetic information in families—a review of guidelines and position papers. Eur J Hum Genet 15: 612–618, 2007. Forrester MB, Merz RD. Epidemiology of triploidy in a population-based birth defects registry, Hawaii, 1986–1999. Am J Med Genet 119A : 319–323, 2003a. Forrester MB, Merz RD. Maternal age-specific Down syndrome rates by maternal race/ethnicity, Hawaii, 1986–2000. Birth Defects Res 67: 625–629, 2003b. Forrester S, Kovach MJ, Smith RE, Rimer L, Wesson M, Kimonis VE. Kousseff syndrome caused by deletion of chromosome 22q11–13. Am J Med Genet 112: 338–342, 2002. Foudila T, Söderström-Anttila V, Hovatta O. Turner’s syndrome and pregnancies after oocyte donation. Hum Reprod 14: 532–535, 1999. Fragouli E, Lenzi M, Ross R , Katz-Jaffe M, Schoolcraft WB, Wells D. Comprehensive molecular cytogenetic analysis of the human blastocyst stage. Hum Reprod 23: 2596–2608, 2008. Fragouli E, Wells D, Doshi A , Gotts S, Harper JC, Delhanty JD. Complete cytogenetic investigation of oocytes from a young cancer patient with the use of comparative genomic hybridisation reveals meiotic errors. Prenat Diagn 26: 71–76, 2006a. Fragouli E, Wells D, Thornhill A , et al. Comparative genomic hybridization analysis of human oocytes and polar bodies. Hum Reprod 21: 2319–2328, 2006b. Fraser FC. Does paternal exposure to Agent Orange cause birth defects? Am J Med Genet 149A : 835–836, 2009. Fridman C, Varela MC, Kok F, Diament A , Koiffmann CP. Paternal UPD 15: further genetic and clinical studies in four Angelman syndrome patients. Am J Med Genet 92: 322–327, 2000.
Fridström M, Ährlund-Richter L, Iwarsson E, et al. Clinical outcome of treatment cycles using preimplantation genetic diagnosis for structural chromosomal abnormalities. Prenat Diagn 21: 781–787, 2001. Friedman J, Adam S, Arbour L, et al. Detection of pathogenic copy number variants in children with idiopathic intellectual disability using 500 K SNP array genomic hybridization. BMC Genomics 10: 526, 2009. Friedman JM, Baross A , Delaney AD, et al. Oligonucleotide microarray analysis of genomic imbalance in children with mental retardation. Am J Hum Genet 79: 500–513, 2006. Friedrich U, Caprani M, Niebuhr E, Therkelsen AJ, Jorgensen AL. Extreme variant of the short arm of chromosome 15. Hum Genet 97: 710–713, 1996. Friedrich U, Nielsen J. Presumptive Y-15 and Y-22 translocation in two families. Hereditas 71: 339–342, 1972. Frints SGM, Schoenmakers EFPM, Smeets E, Petit P, Fryns JP. De novo 7q36 deletion: breakpoint analysis and types of holoprosencephaly. Am J Med Genet 75: 153–158, 1998. Fritz B, Aslan M, Kalscheuer V, et al. Low incidence of UPD in spontaneous abortions beyond the 5th gestational week . Eur J Hum Genet 9: 910–916, 2001. Fritz B, Greber-Platzer S, Frischer T, et al. Familial cryptic translocation with del 4q34→qter and dup 12pter→p13 in sibs with tracheal stenosis: clinical, classical and molecular cytogenetic studies and CGH analyses from archival placental tissues evidencing tertiary trisomy 4 in one abortion specimen. Am J Med Genet 94: 271–280, 2000. Froyen G, Corbett M, Vandewalle J, et al. Submicroscopic duplications of the hydroxysteroid dehydrogenase HSD17B10 and the E3 ubiquitin ligase HUWE1 are associated with mental retardation. Am J Hum Genet 82: 432–443, 2008. Fryburg JS, Dimaio MS, Yang-Feng TL, Mahoney MJ. Follow-up of pregnancies complicated by placental mosaicism diagnosed by chorionic villus sampling. Prenat Diagn 13: 481–494, 1993. Fryns JP, Bulcke J, Hens L, Van den Berghe H. Balanced transmission of centromeric fission products in man. Hum Genet 54: 127–128, 1980. Fryns JP, Kleczkowska A . Reciprocal translocation mosaicism in man. Am J Med Genet 25: 175–176, 1986. Fryns JP, Kleczkowska A . Ring chromosome 21 in the mother and 21/21 translocation in the fetus: karyotype: 45,XX,-21,-21,+t(21;21)(p11;q11). Ann Génét 30: 109–110, 1987.
REFERENCES
•
543
Fryns JP, Kleczkowska A , Kubień E, Van den Berghe H. On the excess of mental retardation and/or congenital malformations in apparently balanced reciprocal translocations. A critical review of the Leuven data 1966–1991. Genet Couns 2: 185–194, 1992a. Fryns JP, Kleczkowska A , Kubien E, Van den Berghe H. XYY syndrome and other Y chromosome polysomies. Mental status and psychosocial functioning. Genet Couns 6: 197–206, 1995. Fryns JP, Kleczkowska A , Limbos C, Vandecasseye W, Van den Berghe H. Centric fission of chromosome 7 with 47,XX,del(7)(pter→cen::q21→qter)+cen fr karyotype in a mother and proximal 7q deletion in two malformed newborns. Ann Génét 28: 248–250, 1985. Fryns JP, Kleczkowska A , Smeets E, van den Berghe H. A new centromeric heteromorphism in the short arm of chromosome 20. J Med Genet 25: 636–637, 1988. Fryns JP, Kleczkowska A, Smeets E, Van Den Berghe H. Transmission of ring chromosome 18 46,XX/46,XX,r(18) mosaicism in a mother and ring chromosome 18 syndrome in her son. Ann Génét 35: 121–123, 1992b. Fryns JP, Petit P. Population cytogenetics of autosomal fragile sites. Clin Genet 31: 61–62, 1987. Fryns JP, Van den Berghe H, Schrander-Stumpel C. Kabuki (Niikawa-Kuroki) syndrome and paracentric inversion of the short arm of chromosome 4. Am J Med Genet 53: 204–205, 1994. Fujimoto S, Yokochi K , Morikawa H, et al. Recurrent cerebral infarctions and del(10)(p14p15.1) de novo in HDR (hypoparathyroidism, sensorineural deafness, renal dysplasia) syndrome. Am J Med Genet 86: 427–429, 1999. Fukunaga M. Immunohistochemical characterization of p57Kip2 expression in tetraploid hydropic placentas. Arch Pathol Lab Med 128: 897–900, 2004. Fullerton G, Hamilton M, Maheshwari A . Should non-mosaic Klinefelter syndrome men be labelled as infertile in 2009? Hum Reprod 25: 588–597, 2010. Fuster C, Miguez L, Miró R , Rigola MA , Perez A , Egozcue J. Familial complex chromosome rearrangement ascertained by in situ hybridisation. J Med Genet 34: 164–166, 1997. Futaki M, Liu JM. Chromosomal breakage syndromes and the BRCA1 genome surveillance complex. Trends Mol Med 7: 560–565, 2001. Gabbett MT, Peters GB, Carmichael JM, Darmanian AP, Collins FA . Prader-Willi syndrome phenocopy due to duplication of Xq21.1-q21.31, with array
544
•
REFERENCES
CGH of the critical region. Clin Genet 73: 353–359, 2008. Gabriel-Robez O, Rumpler Y. The meiotic pairing behaviour in human spermatocytes carrier of chromosome anomalies and their repercussions on reproductive fitness. I: Inversions and insertions. A European collaborative study. Ann Génét 37: 3–10, 1994. Gabriel-Robez O, Ratomponirina C, Dutrillaux B, Carré-Pigeon F, Rumpler Y. Meiotic association between the XY chromosomes and the autosomal quadrivalent of a reciprocal translocation in two infertile men, 46,XY,t(19;22) and 46,XY,t(17;21). Cytogenet Cell Genet 43: 154–160, 1986. Gabriel-Robez O, Rumpler Y, Ratomponirina C, et al. Deletion of the pseudoautosomal region and lack of sex-chromosome pairing at pachytene in two infertile men carrying an X;Y translocation. Cytogenet Cell Genet 54: 38–42, 1990. Gadow EC, Lippold S, Otano L, Serafin E, Scarpati R , Matayoshi T. Chromosome rearrangements among couples with pregnancy losses and other adverse reproductive outcomes. Am J Med Genet 41: 279–281, 1991. Gair JL, Arbour L, Rupps R , Jiang R , Bruyère H, Robinson WP. Recurrent trisomy 21: four cases in three generations. Clin Genet 68: 430–435, 2005. Gajecka M, Gentles AJ, Tsai A , et al. Unexpected complexity at breakpoint junctions in phenotypically normal individuals and mechanisms involved in generating balanced translocations t(1;22)(p36;q13). Genome Res 18: 1733–1742, 2008a. Gajecka M, Mackay KL, Shaffer LG. Monosomy 1p36 deletion syndrome. Am J Med Genet 145C: 346–356, 2007. Gajecka M, Pavlicek A , Glotzbach CD, et al. Identification of sequence motifs at the breakpoint junctions in three t(1;9)(p36.3;q34) and delineation of mechanisms involved in generating balanced translocations. Hum Genet 120: 519–526, 2006. Gajecka M, Saadeh R , Mackay KL, et al. Clinical and molecular cytogenetic characterization of four patients with unbalanced translocation der(1) t(1;22)(p36;q13). Am J Med Genet 146A : 2777–2784, 2008b. Gajecka M, Saitta SC, Gentles AJ, et al. Recurrent interstitial 1p36 deletions: Evidence for germline mosaicism and complex rearrangement breakpoints. Am J Med Genet 152A : 3074–3083, 2010. Gajecka M, Yu W, Ballif BC, et al. Delineation of mechanisms and regions of dosage imbalance in complex rearrangements of 1p36 leads to a
putative gene for regulation of cranial suture closure. Eur J Hum Genet 13: 139–149, 2005. Galasso C, Lo-Castro A , El-Malhany N, et al. Mild Wolf-Hirschhorn phenotype in a girl with unbalanced t(4p;12p) translocation without seizures. Am J Med Genet 152A : 258–261, 2010. Garchet-Beaudron A , Dreux S, Leporrier N, Oury JF, Muller F. Second-trimester Down syndrome maternal serum marker screening: a prospective study of 11 040 twin pregnancies. Prenat Diagn 28: 1105–1109, 2008. García-Castillo H, Vásquez-Velásquez AI, Rivera H, Barros-Núñez P. Clinical and genetic heterogeneity in patients with mosaic variegated aneuploidy: delineation of clinical subtypes. Am J Med Genet 146A : 1687–1695, 2008. Garcia-Heras J, Martin JA , Witchel SF, Scacheri P. De novo der(X)t(X;10)(q26;q21) with features of distal trisomy 10q: case report of paternal origin identified by late replication with BrdU and the human androgen receptor assay (HAR). J Med Genet 34: 242–245, 1997. Garcia-Heras J, Ulm JE, Shaver DE, Hall E, Saikevych IA . Prenatal diagnosis of a der(X) t(X;15)(p22.2;q11.2) inherited from a maternal translocation X;15. Prenat Diagn 18: 747–750, 1998. Gardner RJM, Dockery HE, Fitzgerald PH, et al. Mosaicism with a normal cell line and an autosomal structural rearrangement. J Med Genet 31: 108–114, 1994. Gardner RJM, Grindley RM, Chewings WE, Clarkson JE. Ring chromosome 1 associated with radial ray defect. J Med Genet 21: 400, 1984. Gardner RJM, Monk NA , Allen GJ, Parslow MI. A three way translocation in mother and daughter. J Med Genet 23: 90, 1986a. Gardner RJM, Monk NA , Clarkson JE, Allen GJ. Ring 21 chromosome: the mild end of the phenotypic spectrum. Clin Genet 30: 466–470, 1986b. Gardner RJM, Rudd NL, Stevens LJ, Worton RG. Autosomal imbalance with a near-normal phenotype: the small effect of trisomy for the short arm of chromosome 18. Birth Defects Orig Artic Ser 14: 359–363, 1978. Gardner RJM, Parslow MI, Veale AMO. The formation of the abnormal chromosome in balanced homologous Robertsonian translocation carriers. Humangenetik 21: 270–282, 1974. Gardner RJM, Veale AMO. De novo translocation Down’s syndrome: risk of recurrence of Down’s syndrome. Clin Genet 6: 160–164, 1974. Gardner RJM, Veale AMO, Parslow MI, et al. A survey of 972 cytogenetically examined cases of Down’s syndrome. NZ Med J 78: 403–409, 1973a.
Gardner RJM, Veale AMO, Sands VE, Holdaway MDH. XXXX syndrome: case report, and a note on genetic counselling and fertility. Humangenetik 17: 323–330, 1973b. Garrett LA , Garner EI, Feltmate CM, Goldstein DP, Berkowitz RS. Subsequent pregnancy outcomes in patients with molar pregnancy and persistent gestational trophoblastic neoplasia. J Reprod Med 53: 481–486, 2008. Gassner R , Metzenbauer M, Hafner E, Vallazza U, Philipp K . Triploidy in a twin pregnancy: small placenta volume as an early sonographical marker. Prenat Diagn 23: 16–20, 2003. Gautier M, Harper PS. Fiftieth anniversary of trisomy 21: returning to a discovery. Hum Genet 126: 317–324, 2009. Gay CT, Hardies LJ, Rauch RA , et al. Magnetic resonance imaging demonstrates incomplete myelination in 18q- syndrome: evidence for myelin basic protein haploinsufficiency. Am J Med Genet 74: 422–431, 1997. Gecz J. The FMR2 gene, FRAXE and non-specific X-linked mental retardation: clinical and molecular aspects. Ann Hum Genet 64: 95–106, 2000. Gecz J, Gedeon AK , Sutherland GR , Mulley JC. Identification of the gene FMR2, associated with FRAXE mental retardation. Nat Genet 13: 105–108, 1996. Geigy Scientific Tables: Introduction to Statistics, Statistical Tables, Mathematical Formulae. Basle, Switzerland: Novartis (formerly Ciba Geigy), 1982. Gekas J, Thepot F, Turleau C, et al. Chromosomal factors of infertility in candidate couples for ICSI: an equal risk of constitutional aberrations in women and men. Hum Reprod 16: 82–90, 2001. Genest P. Transmission héréditaire depuis 300 ans d’un chromosome Y à satellites dans une lignée familiale. Ann Génét 16: 35–38, 1973. Gentile M, Volpe P, Cariola F, et al. Prenatal diagnosis of chromosome 4 mosaicism: prognostic role of cytogenetic, molecular, and ultrasound/MRI characterization. Am J Med Genet 136: 66–70, 2005. Genuardi M, Tozzi C, Pomponi MG, et al. Mosaic trisomy 17 in amniocytes: phenotypic outcome, tissue distribution, and uniparental disomy studies. Eur J Hum Genet 7: 421–426, 1999. George AM, Oei P, Winship I. False-positive diagnosis of trisomy 21 using fluorescence in situ hybridisation (FISH) on uncultured amniotic fluid cells. Prenat Diagn 23: 302–305, 2003. Georgiades P, Chierakul C, Ferguson-Smith AC. Parental origin effects in human trisomy for
REFERENCES
•
545
chromosome 14q: implications for genomic imprinting. J Med Genet 35: 821–824, 1998. Geraedts J, Collins J, Gianaroli L, et al. What next for preimplantation genetic screening? A polar body approach! Hum Reprod 25: 575–577, 2010. Gerkes EH, van der Kevie-Kersemaekers AM, Yakin M, Smeets DFCM, van Ravenswaaij-Arts CMA . The importance of chromosome studies in Roberts syndrome/SC phocomelia and other cohesinopathies. Eur J Med Genet 53: 40–44, 2010. German J. Bloom syndrome: a mendelian prototype of somatic mutational disease. Medicine (Baltimore) 72: 393–406, 1993. German J, Ellis NA . Bloom syndrome. In: The Online Metabolic and Molecular Bases of Inherited Disease, www.ommbid.com, 2011, Chapter 30. German J, Sanz MM, Ciocci S, Ye TZ, Ellis NA . Syndrome-causing mutations of the BLM gene in persons in the Bloom’s Syndrome Registry. Hum Mutat 28: 743–753, 2007. Ghosh A , Higgins L, Larkins SA , et al. Prenatal diagnosis and prenatal imaging of a de novo 46,X,der(Y)t(X;Y)(p22.13;q11.23) leading to functional disomy for the distal end of the X chromosome short arm from Xp22.13 in a phenotypically male fetus with posterior fossa abnormalities. Prenat Diagn 28: 1068–1071, 2008. Ghosh S, Feingold E, Dey SK . Etiology of Down syndrome: evidence for consistent association among altered meiotic recombination, nondisjunction, and maternal age across populations. Am J Med Genet 149A : 1415–1420, 2009. Giacalone JP, Francke U. Common sequence motifs at the rearrangement sites of a constitutional X/ autosome translocation and associated deletion. Am J Hum Genet 50: 725–741, 1992. Giampietro PF, Verlander PC, Davis JG, Auerbach AD. Diagnosis of Fanconi anemia in patients without congenital malformations: an international Fanconi Anemia Registry Study. Am J Med Genet 68: 58–61, 1997. Gianaroli L, Magli MC, Cavallini G, et al. Frequency of aneuploidy in sperm from patients with extremely severe male factor infertility. Hum Reprod 20: 2140–2152, 2005. Gianaroli L, Magli MC, Ferraretti AP, et al. Possible interchromosomal effect in embryos generated by gametes from translocation carriers. Hum Reprod 17: 3201–3207, 2002. Giardino D, Corti C, Ballarati L, et al. Prenatal diagnosis of a de novo complex chromosome rearrangement (CCR) mediated by six breakpoints, and a review of 20 prenatally
546
•
REFERENCES
ascertained CCRs. Prenat Diagn 26: 565–570, 2006. Giardino D, Corti C, Ballarati L, et al. De novo balanced chromosome rearrangements in prenatal diagnosis. Prenat Diagn 29: 257–265, 2009. Giardino D, Finelli P, Russo S, et al. Small familial supernumerary ring chromosome 2: FISH characterization and genotype-phenotype correlation. Am J Med Genet 111: 319–323, 2002. Gibbons B, Tan SY, Yu CC, Cheah E, Tan HL. Risk of gonadoblastoma in female patients with Y chromosome abnormalities and dysgenetic gonads. J Paediatr Child Health 35: 210–213, 1999. Gibson LH, McGrath J, Yang-Feng TL. A complex chromosome rearrangement with at least five breakpoints studied by fluorescence in situ hybridization. Am J Med Genet 68: 417–420, 1997. Gibson WT, Harvard C, Qiao Y, Somerville MJ, Lewis ME, Rajcan-Separovic E. Phenotypegenotype characterization of alpha-thalassemia mental retardation syndrome due to isolated monosomy of 16p13.3. Am J Med Genet 146A : 225–232, 2008. Gidoni YS, Takefman J, Holzer HE, et al. Cryopreservation of a mother’s oocytes for possible future use by her daughter with Turner syndrome: case report. Fertil Steril 90: 2008 e9–12, 2008. Giglio S, Calvari V, Gregato G, et al. Heterozygous submicroscopic inversions involving olfactory receptor-gene clusters mediate the recurrent t(4;8) (p16;p23) translocation. Am J Hum Genet 71: 276–285, 2002. Giglio S, Pirola B, Arrigo G, et al. Opposite deletions/ duplications of the X chromosome: two novel reciprocal rearrangements. Eur J Hum Genet 8: 63–70, 2000. Gilardi JL, Perrotin F, Paillet C, et al. Prenatal diagnosis of trisomy 21 by i(21q): a rare case of fetoplacental chromosomal discrepancy. Prenat Diagn 22: 856–858, 2002. Gilbert B, Yardin C, Briault S, et al. Prenatal diagnosis of female monozygotic twins discordant for Turner syndrome: implications for prenatal genetic counselling. Prenat Diagn 22: 697–702, 2002. Gilgenkrantz S, Cabrol C, Lausecker C, Hartleyb ME, Bohe B. Le syndrome Dr: étude d’un nouveau cas (46,XX,14r). Ann Génét 14: 23–31, 1971. Gilling M, Dullinger JS, Gesk S, et al. Breakpoint cloning and haplotype analysis indicate a single origin of the common Inv(10)(p11.2q21.2) mutation among northern Europeans. Am J Hum Genet 78: 878–883, 2006. Gillon R . On sterilising severely mentally handicapped people. J Med Ethics 13: 59–61, 1987.
Giltay JC, Kastrop PM, Tiemessen CH, van Inzen WG, Scheres JM, Pearson PL. Sperm analysis in a subfertile male with a Y;16 translocation, using four-color FISH. Cytogenet Cell Genet 84: 67–72, 1999. Giltay JC, van Golde RJT, Kastrop PMM. Analysis of spermatozoa from seven ICSI males with constitutional sex chromosomal abnormalities by fluorescent in situ hybridization. J Assist Reprod Genet 17: 151–155, 2000. Gimelbrant A , Hutchinson JN, Thompson BR , Chess A . Widespread monoallelic expression on human autosomes. Science 318: 1136–1140, 2007. Gimelli G, Giorda R , Beri S, Gimelli S, Zuffardi O. A 46,X,inv(Y) young woman with gonadal dysgenesis and gonadoblastoma: cytogenetics, molecular, and methylation studies. Am J Med Genet 140: 40–45, 2006. Ginsburg C, Fokstuen S, Schinzel A . The contribution of uniparental disomy to congenital development defects in children born to mothers at advanced childbearing age. Am J Med Genet 95: 454–460, 2000. Ginsberg NA , Cadkin A , Strom C, Bauer-Marsh E, Verlinsky Y. Prenatal diagnosis of 46,XX male fetuses. Am J Obstet Gynecol 180: 1006–1007, 1999. Giorda R , Bonaglia MC, Beri S, et al. Complex segmental duplications mediate a recurrent dup(X)(p11.22-p11.23) associated with mental retardation, speech delay, and EEG anomalies in males and females. Am J Hum Genet 85: 394–400, 2009. Giorda R , Bonaglia MC, Milani G, et al. Molecular and cytogenetic analysis of the spreading of X inactivation in a girl with microcephaly, mild dysmorphic features and t(X;5)(q22.1;q31.1). Eur J Hum Genet 16: 897–905, 2008. Giorda R , Cerritello A , Bonaglia MC, et al. Selective disruption of muscle and brain-specific BPAG1 isoforms in a girl with a 6;15 translocation, cognitive and motor delay, and tracheooesophageal atresia. J Med Genet 41: e71, 2004. Giraudeau F, Taine L, Biancalana V, et al. Use of a set of highly polymorphic minisatellite probes for the identification of cryptic 1p36.3 deletions in a large collection of patients with idiopathic mental retardation. J Med Genet 38: 121–125, 2001. Girirajan S, Rosenfeld JA , Cooper GM, et al. A recurrent 16p12.1 microdeletion supports a two-hit model for severe developmental delay. Nat Genet 42: 203–209, 2010. Girirajan S, Truong HT, Blanchard CL, Elsea SH. A functional network module for Smith-Magenis syndrome. Clin Genet 75: 364–374, 2009.
Glass IA , Rauen KA , Chen E, et al. Ring chromosome 15: characterization by array CGH. Hum Genet 118: 611–617, 2006. Glass IA , Stormer P, Oei PT, Hacking E, Cotter PD. Trisomy 2q11.2→q21.1 resulting from an unbalanced insertion in two generations. J Med Genet 35: 319–322, 1998. Glasson EJ, Sullivan SG, Hussain R , Petterson BA , Montgomery PD, Bittles AH. The changing survival profile of people with Down’s syndrome: implications for genetic counselling. Clin Genet 62: 390–393, 2002. Gleeson JG, Allen KM, Fox JW, et al. Doublecortin, a brain-specific gene mutated in human X-linked lissencephaly and double cortex syndrome, encodes a putative signaling protein. Cell 92: 63–72, 1998. Gleicher N, Weghofer A , Barad DH. A pilot study of premature ovarian senescence: I. Correlation of triple CGG repeats on the FMR1 gene to ovarian reserve parameters FSH and anti-Müllerian hormone. Fertil Steril 91: 1700–1706, 2009. Glenn CC, Deng G, Michaelis RC, et al. DNA methylation analysis with respect to prenatal diagnosis of the Angelman and Prader-Willi syndromes and imprinting. Prenat Diagn 20: 300–306, 2000. Gokden M, Al-Hinti JT, Harik SI. Peripheral nervous system pathology in fragile X tremor/ataxia syndrome (FXTAS). Neuropathol 29: 280–284, 2009. Golden WL, Sudduth KW, Burnett SH, Kelly TE. Mosaicism in Prader-Willi syndrome: detection using fluorescent in situ hybridization. Am J Med Genet 85: 424–425, 1999. Goldman ASH, Hultén MA . Chromosome in situ suppression hybridisation in human male meiosis. J Med Genet 29: 98–102, 1992. Goldman ASH, Martin RH, Johannisson R , et al. Meiotic and sperm chromosome analysis in a male carrier of an inverted insertion (3;10) (q13.2;p14p13). J Med Genet 29: 460–464, 1992. Gole L, Crolla JA , Thomas SN, Jacobs PA , Dennis NR . Characterization of breakpoints in the GABRG3 and TSPY genes in a family with a t(Y;15)(p11.2;q12). Am J Med Genet 125A : 177–180, 2004. Gole LA , Biswas A . Characterization of a small supernumerary marker chromosome as r(8) at prenatal diagnosis by MFISH. Prenat Diagn 25: 73–78, 2005. Golubovsky MD. Postzygotic diploidization of triploids as a source of unusual cases of mosaicism, chimerism and twinning. Hum Reprod 18: 236–242, 2003.
REFERENCES
•
547
Golzio C, Guirchoun J, Ozilou C, et al. Cytogenetic and histological features of a human embryo with homogeneous chromosome 8 trisomy. Prenat Diagn 26: 1201–1205, 2006. Goodall J. Helping a child to understand her own testicular feminisation. Lancet 337: 33–35, 1991. Goodwin PJ, Ennis M, Pritchard KI, Trudeau M, Hood N. Risk of menopause during the first year after breast cancer diagnosis. J Clin Oncol 17: 2365–2370, 1999. Goossens V, Harton G, Moutou C, et al. ESHRE PGD Consortium data collection VIII: cycles from January to December 2005 with pregnancy follow-up to October 2006. Hum Reprod 23: 2629–2645, 2008. Goumy C, Beaufrère AM, Francannet C, et al. Prenatal detection of mosaic isochromosome 20q: a fourth report with abnormal phenotype. Prenat Diagn 25: 653–655, 2005. Gradek GA , Kvistad PH, Houge G. Monosomy 8 rescue gave cells with a normal karyotype in a mildly affected man with 46,XY,r(8) mosaicism. Eur J Med Genet 49: 292–297, 2006. Grams SE, R and L, Norton ME. Complete isochromosome 5p in one fetus of a monochorionic twin pair. Prenat Diagn 31: 605–607, 2011. Grange DK , Garcia-Heras J, Kilani RA , Lamp S. Trisomy 20q13→20qter in a girl with multiple congenital malformations and a recombinant chromosome 20 inherited from a paternal inversion (20)(p13q13.1): clinical report and review of the trisomy 20q phenotype. Am J Med Genet 137A : 308–312, 2005. Grass FS, Schwartz RP, Deal JO, Parke JC, Jr. Gonadal dysgenesis, intra-X chromosome insertion, and possible position effect in an otherwise normal female. Clin Genet 20: 28–35, 1981. Grati FR , Barlocco A , Grimi B, et al. Chromosome abnormalities investigated by non-invasive prenatal testing account for approximately 50% of fetal unbalances associated with relevant clinical phenotypes. Am J Med Genet 152A : 1434–1442, 2010. Grati FR , Turolla L, D’Ajello P, et al. Chromosome 11 segmental paternal isodisomy in amniocytes from two fetuses with omphalocoele: new highlights on phenotype-genotype correlations in BeckwithWiedemann syndrome. J Med Genet 44: 257–263, 2007. Gravholt CH. Medical problems of adult Turner’s syndrome. Horm Res 56(suppl 1): 44–50, 2001. Gravholt CH, Weis Naeraa R . Reference values for body proportions and body composition in adult
548
•
REFERENCES
women with Ullrich-Turner syndrome. Am J Med Genet 72: 403–408, 1997. Greally JM, Neiswanger K , Cummins JH, et al. A molecular anatomical analysis of mosaic trisomy 16. Hum Genet 98: 86–90, 1996. Greco CM, Hagerman RJ, Tassone F, et al. Neuronal intranuclear inclusions in a new cerebellar tremor/ ataxia syndrome among fragile X carriers. Brain 125: 1760–1771, 2002. Green DM, Kawashima T, Stovall M, et al. Fertility of female survivors of childhood cancer: a report from the childhood cancer survivor study. J Clin Oncol 27: 2677–2685, 2009. Green DM, Kawashima T, Stovall M, et al. Fertility of male survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J Clin Oncol 28: 332–339, 2010. Green JM, Hewison J, Bekker HL, Bryant LD, Cuckle HS. Psychosocial aspects of genetic screening of pregnant women and newborns: a systematic review. Health Technol Assess (UK) 8(33): 1–124, 2004. Greger V, Knoll JHM, Wagstaff J, et al. Angelman syndrome associated with an inversion of chromosome 15q11.2q24.3. Am J Hum Genet 60: 574–580, 1997. Gregg C, Zhang J, Weissbourd B, et al. Highresolution analysis of parent-of-origin allelic expression in the mouse brain. Science 329: 643–648, 2010. Gribble SM, Fiegler H, Burford DC, et al. Applications of combined DNA microarray and chromosome sorting technologies. Chromosome Res 12: 35–43, 2004. Griffin DK , Hyland P, Tempest HG, Homa ST. Safety issues in assisted reproduction technology: Should men undergoing ICSI be screened for chromosome abnormalities in their sperm? Hum Reprod 18: 229–235, 2003. Griffiths MJ, Miller PR , Stibbe HM. A false-positive diagnosis of Turner syndrome by amniocentesis. Prenat Diagn 16: 463–466, 1996. Grigorescu-Sido A , Heinrich U, Grigorescu-Sido P, et al. Three new 46,XX male patients: a clinical, cytogenetic and molecular analysis. J Pediatr Endocrinol Metab 18: 197–203, 2005. Grillo L, Reitano S, Belfiore G, et al. Familial 1.1 Mb deletion in chromosome Xq22.1 associated with mental retardation and behavioural disorders in female patients. Eur J Med Genet 53: 113–116, 2010. Grisart B, Willatt L, Destrée A , et al. 17q21.31 microduplication patients are characterised by behavioural problems and poor social interaction. J Med Genet 46: 524–530, 2009.
Grobman WA , Auger M, Shulman LP, Elias S. The association between chorionic villus sampling and preeclampsia. Prenat Diagn 29: 800–803, 2009. Groen SE, Drewes JG, de Boer EG, Hoovers JMN, Hennekam RCM. Repeated unbalanced offspring due to a familial translocation involving chromosomes 5 and 6. Am J Med Genet 80: 448–453, 1998. Grønskov K , Hallberg A , Brøndum-Nielsen K . Mutational analysis of the FMR1 gene in 118 mentally retarded males suspected of fragile X syndrome: absence of prevalent mutations. Hum Genet 102: 440–445, 1998. Grønskov K , Brøndum-Nielsen K , Dedic A , Hjalgrim H. A nonsense mutation in FMR1 causing fragile X syndrome. Eur J Hum Genet 19: 489–491, 2011. Gross M, Hanenberg H, Lobitz S, et al. Reverse mosaicism in Fanconi anemia: natural gene therapy via molecular self-correction. Cytogenet Genome Res 98: 126–135, 2002. Gross SJ, Tharapel AT, Phillips OP, Shulman LP, Pivnick EK , Park VM. A jumping Robertsonian translocation: a molecular and cytogenetic study. Hum Genet 98: 291–296, 1996. Grossfeld PD, Mattina T, Lai Z, et al. The 11q terminal deletion disorder: a prospective study of 110 cases. Am J Med Genet 129A : 51–61, 2004. Grossmann V, Höckner M, Karmous-Benailly H, et al. Parental origin of apparently balanced de novo complex chromosomal rearrangements investigated by microdissection, whole genome amplification, and microsatellite-mediated haplotype analysis. Clin Genet 78: 548–553, 2010. Groupe de Cytogénéticiens Français. Pericentric inversions in man. A French collaborative study. Ann Génét 29: 129–168, 1986a. Groupe de Cytogénéticiens Français. Paracentric inversions in man. A French collaborative study Ann Génét 29: 169–176, 1986b. Grover S, Brady S, Chondros P. Sterilisations in girls and young women: is it still happening? Aust NZ J Public Health 26: 273–275, 2002. Gruchy N, Barreau M, Kessler K , Gourdier D, Leporrier N. A paternally transmitted complex chromosomal rearrangement (CCR) involving chromosomes 2, 6, and 18 includes eight breakpoints and five insertional translocations (ITs) through three generations. Am J Med Genet 152A : 185–190, 2010. Gruchy N, Lebrun M, Herlicoviez M, et al. Supernumerary marker chromosomes management in prenatal diagnosis. Am J Med Genet 146A : 2770–2776, 2008.
Gu W, Lupski JR . CNV and nervous system diseases—what’s new? Cytogenet Genome Res 123: 54–64, 2008. Gu W, Zhang F, Lupski JR . Mechanisms for human genomic rearrangements. Pathogenet 1: 4, 2008. Guanciali-Franchi P, Calabrese G, Morizio E, et al. Identification of 14 rare marker chromosomes and derivatives by spectral karyotyping in prenatal and postnatal diagnosis. Am J Med Genet 127A : 144–148, 2004. Guc-Scekic M, Milasin J, Stevanovic M, Stojanov LJ, Djordjevic M. Tetraploidy in a 26-month-old girl (cytogenetic and molecular studies). Clin Genet 61: 62–65, 2002. Guediche N, Brisset S, Benichou JJ, et al. Chromosomal breakpoints characterization of two supernumerary ring chromosomes 20. Am J Med Genet 152A : 464–471, 2010. Guichaoua MR , Devictor M, Hartung M, Luciani JM, Stahl A . Random acrocentric bivalent associations in human pachytene spermatocytes. Molecular implications in the occurrence of Robertsonian translocations. Cytogenet Cell Genet 42: 191–197, 1986. Guichaoua MR , Quack B, Speed RM, Noel B, Chandley AC, Luciani JM. Infertility in human males with autosomal translocations: meiotic study of a 14;22 Robertsonian translocation. Hum Genet 86: 162–166, 1990. Guioli S, Incerti B, Zanaria E, et al. Kallmann syndrome due to a translocation resulting in an X/Y fusion gene. Nat Genet 1: 337–340, 1992. Gunay-Aygun M, Heeger S, Schwartz S, Cassidy SB. Delayed diagnosis in patients with Prader-Willi syndrome due to maternal uniparental disomy 15. Am J Med Genet 71: 106–110, 1997. Gunn SR , Mohammed M, Reveles XT, et al. Molecular characterization of a patient with central nervous system dysmyelination and cryptic unbalanced translocation between chromosomes 4q and 18q. Am J Med Genet 120A : 127–135, 2003. Gunnarsson C, Graffmann B, Jonasson J. Chromosome r(10)(p15.3q26.12) in a newborn child: case report. Mol Cytogenet 2: 25, 2009. Gurrieri F, Battaglia A , Torrisi L, et al. Pervasive developmental disorder and epilepsy due to maternally derived duplication of 15q11-q13. Neurology 52: 1694–1697, 1999. Gustashaw KM, Zurcher V, Dickerman LH, Stallard R , Willard HF. Partial X chromosome trisomy with functional disomy of Xp due to failure of X inactivation. Am J Med Genet 53: 39–45, 1994. Guttenbach M, Engel W, Schmid M. Analysis of structural and numerical chromosome
REFERENCES
•
549
abnormalities in sperm of normal men and carriers of constitutional chromosome aberrations. A review. Hum Genet 100: 1–21, 1997. Guttenbach M, Kohn FM, Engel W, Schmid M. Meiotic nondisjunction of chromosomes 1, 17, 18, X, and Y in men more than 80 years of Age. Biol Reprod 63: 1727–1729, 2000. Guttenbach M, Koschorz B, Bernthaler U, Grimm T, Schmid M. Sex chromosome loss and aging: in situ hybridization studies on human interphase nuclei. Am J Hum Genet 57: 1143–1150, 1995. Guzé C, Qin N, Kelly J, et al. Isochromosome 22 in trisomy 22 mosaic with five cell lines. Am J Med Genet 124A : 79–84, 2004. Gyejye A , Anyane-Yeboa K , Warburton D. Follow-up of 30 children with de novo balanced rearrangements and supernumerary marker chromosomes diagnosed at amniocentesis. Am J Hum Genet 69: 208, 2001. Haas BW, Barnea-Goraly N, Lightbody AA , et al. Early white-matter abnormalities of the ventral frontostriatal pathway in fragile X syndrome. Dev Med Child Neurol 51: 593–599, 2009. Habecker-Green J, Naeem R , Goh W, Pflueger S, Murray M, Cohn G. Reproduction in a patient with trisomy 8 mosaicism: case report and literature review. Am J Med Genet 75: 382–385, 1998. Hadjiathanasiou CG, Brauner R , Lortat-Jacob S, et al. True hermaphroditism: genetic variants and clinical management. J Pediatr 125: 738–744, 1994. Hagerman RJ, Berry-Kravis E, Kaufmann WE, et al. Advances in the treatment of fragile X syndrome. Pediatrics 123: 378–390, 2009. Hagerman RJ, Leehey M, Heinrichs W, et al. Intention tremor, parkinsonism, and generalized brain atrophy in male carriers of fragile X . Neurology 57: 127–130, 2001. Hagleitner MM, Lankester A , Maraschio P, et al. Clinical spectrum of immunodeficiency, centromeric instability and facial dysmorphism (ICF syndrome). J Med Genet 45: 93–99, 2008. Hahnemann JM, Nir M, Friberg M, Engel U, Bugge M. Trisomy 10 mosaicism and maternal uniparental disomy 10 in a liveborn infant with severe congenital malformations. Am J Med Genet 138A : 150–154, 2005. Hahnemann JM, Vejerslev LO. European collaborative research on mosaicism in CVS (EUCROMIC)— fetal and extrafetal cell lineages in 192 gestations with CVS mosaicism involving single autosomal trisomy. Am J Med Genet 70: 179–187, 1997.
550
•
REFERENCES
Hajianpour A , Murer-Orlando M, Docherty Z. Germ line mosaicism for chromosome 5 “cri-du-chat” deletion? Am J Hum Genet 49: 217, 1991. Hale DW. Is X-Y recombination necessary for spermatocyte survival during mammalian spermatogenesis? Cytogenet Cell Genet 65: 278–282, 1994. Hall BD. Recurrence risk in de novo 21q21q translocation Down syndrome. Am J Med Genet 22: 417–418, 1985. Hall BD, Cadle RG. Adjunct diagnostic test for Angelman syndrome: the tuning fork response. Am J Med Genet 112: 429, 2002. Halliday J, Oke K , Breheny S, Algar E, Amor DJ. Beckwith-Wiedemann syndrome and IVF: a case-control study. Am J Hum Genet 75: 526–528, 2004. Halliday JL, Ukoumunne OC, Baker HWG, et al. Increased risk of blastogenesis birth defects, arising in the first 4 weeks of pregnancy, after assisted reproductive technologies. Hum Reprod 25: 59–65, 2010. Halliday JL, Warren R , McDonald G, Rice PL, Bell RJ, Watson LF. Prenatal diagnosis for women aged 37 years and over: to have or not to have. Prenat Diagn 21: 842–847, 2001. Halliday JL, Watson LF, Lumley J, Danks DM, Sheffield LJ. New estimates of Down syndrome risks at chorionic villus sampling, amniocentesis, and livebirth in women of advanced maternal age from a uniquely defined population. Prenat Diagn 15: 455–465, 1995. Hamel BCJ, Smits APT, de Graaff E, et al. Segregation of FRAXE in a large family: clinical, psychometric, cytogenetic, and molecular data. Am J Hum Genet 55: 923–931, 1994. Hammond LS, Macias MM, Tarleton JC, Shashidhar Pai G. Fragile X syndrome and deletions in FMR 1: new case and review of the literature. Am J Med Genet 72: 430–434, 1997. Hammoud I, Molina Gomes D, Bergere M, Wainer R , Selva J, Vialard F. Sperm chromosome analysis of an infertile patient with a 95% mosaic r(21) karyotype and normal phenotype. Fertil Steril 91: 930 e13–15, 2009. Han JY, Shin JH, Han MS, Je GH, Shaffer LG. Microarray detection of a de novo der(X)t(X;11) (q28;p13) in a girl with premature ovarian failure and features of Beckwith-Wiedemann syndrome. J Hum Genet 51: 641–643, 2006. Hande MP, Azizova TV, Geard CR , et al. Past exposure to densely ionizing radiation leaves a unique permanent signature in the genome. Am J Hum Genet 72: 1162–1170, 2003.
Hani VH, Park J, Allen EF, Moeschler JB. Paracentric inversion of chromosome 18 with duplication of the inverted material in a child due to maternal paracentric inversion of 18q. Am J Hum Genet 57: A115, 1995. Hannes FD, Sharp AJ, Mefford HC, et al. Recurrent reciprocal deletions and duplications of 16p13. 11: the deletion is a risk factor for MR/MCA while the duplication may be a rare benign variant. J Med Genet 46: 223–232, 2009. Hanson C, Hardarson T, Lundin K , et al. Re-analysis of 166 embryos not transferred after PGS with advanced reproductive maternal age as indication. Hum Reprod 24: 2960–2964, 2009. Happle R , König A . Dominant traits may give rise to paired patches of either excessive or absent involvement. Am J Med Genet 84: 176–177, 1999. Hardy K , Wright C, Rice S, et al. Future developments in assisted reproduction in humans. Reproduction 123: 171–183, 2002. Härkönen K , Viitanen T, Larsen SB, Bonde JP, Lähdetie J. Aneuploidy in sperm and exposure to fungicides and lifestyle factors. ASCLEPIOS. A European Concerted Action on Occupational Hazards to Male Reproductive Capability. Environ Mol Mutagen 34: 39–46, 1999. Harmon RJ, Bender BG, Linden MG, Robinson A . Transition from adolescence to early adulthood: adaptation and psychiatric status of women with 47,XXX . J Am Acad Child Adolesc Psychiatry 37: 286–291, 1998. Harper J, Coonen E, De Rycke M, et al. What next for preimplantation genetic screening (PGS)? A position statement from the ESHRE PGD Consortium Steering Committee. Hum Reprod 25: 821–823, 2010a. Harper JC, Coonen E, De Rycke M, et al. ESHRE PGD Consortium data collection X: cycles from January to December 2007 with pregnancy follow-up to October 2008. Hum Reprod 25: 2685–2707, 2010b. Harper PS. The discovery of the human chromosome number in Lund, 1955–1956. Hum Genet 119: 226–232, 2006. Harper PS. First years of human chromosomes. The beginnings of human cytogenetics. Bloxham, England: Scion Publishing , 2006. Harper PS. Practical Genetic Counseling. 7th ed. New York: Oxford University Press, 2010. Harris DJ, Hankins L, Begleiter ML. Reproductive risk of t(13q14q) carriers: case report and review. Am J Med Genet 3: 175–181, 1979. Harrison KJ, Barrett IJ, Lomax BL, Kuchinka BD, Kalousek DK . Detection of confined placental mosaicism in trisomy 18 conceptions using
interphase cytogenetic analysis. Hum Genet 92: 353–358, 1993. Hasegawa T, Harada N, Ikeda K , et al. Digynic triploid infant surviving for 46 days. Am J Med Genet 87: 306–310, 1999. Hashish AF, Monk NA , Watt AJ, Gardner RJM. A de novo insertion, detected prenatally, with normal phenotype. J Med Genet 29: 351, 1992. Hassan NH. Miconazole genotoxicity in mice. J Appl Toxicol 17: 313–319, 1997. Hassold T, Arnovitz K , Jacobs PA , May K , Robinson D. The parental origin of the missing or additional chromosome in 45,X and 47,XXX females. Birth Defects Orig Artic Ser 26: 297–304, 1990a. Hassold T, Hunt PA , Sherman S. Trisomy in humans: incidence, origin and etiology. Curr Opin Genet Dev 3: 398–403, 1993. Hassold T, Jacobs PA , Pettay D. Analysis of nucleolar organizing regions in parents of trisomic spontaneous abortions. Hum Genet 76: 381–384, 1987. Hassold T, Pettay D, May K , Robinson A . Analysis of non-disjunction in sex chromosome tetrasomy and pentasomy. Hum Genet 85: 648–650, 1990b. Hassold T, Sherman S. Down syndrome: genetic recombination and the origin of the extra chromosome 21. Clin Genet 57: 95–100, 2000. Hassold TJ. Nondisjunction in the human male. Curr Top Dev Biol 37: 383–406, 1998. Hastings RJ, Nisbet DL, Waters K , Spencer T, Chitty LS. Prenatal detection of extra structurally abnormal chromosomes (ESACs): new cases and a review of the literature. Prenat Diagn 19: 436–445, 1999a. Hastings RJ, Watson SG, Chitty LS. Prenatal finding of a fetus with mosaicism for two balanced de novo chromosome rearrangements. Prenat Diagn 19: 77–80, 1999b. Hatakeyama C, Gao H, Harmer K , Ma S. Meiotic segregation patterns and ICSI pregnancy outcome of a rare (13;21) Robertsonian translocation carrier: a case report. Hum Reprod 21: 976–979, 2006. Hatchwell E, Robinson D, Crolla JA , Cockwell AE. X inactivation analysis in a female with hypomelanosis of Ito associated with a balanced X;17 translocation: evidence for functional disomy of Xp. J Med Genet 33: 216–220, 1996. Hattori M, Fujiyama A , Taylor TD, et al. The DNA sequence of human chromosome 21. Nature 405: 311–319, 2000. Hatziioannou AG, Krauss CM, Lewis MB, Halazonetis TD. Familial holoprosencephaly associated with a translocation breakpoint at
REFERENCES
•
551
chromosomal position 7q36. Am J Med Genet 40: 201–205, 1991. Haverty CE, Lin AE, Simpson E, Spence MA , Martin RA . 47,XXX associated with malformations. Am J Med Genet 125A : 108–111, 2004. Havlovicova M, Novotna D, Kocarek E, et al. A girl with neurofibromatosis type 1, atypical autism and mosaic ring chromosome 17. Am J Med Genet 143: 76–81, 2007. Hayward BE, De Vos M, Talati N, et al. Genetic and epigenetic analysis of recurrent hydatidiform mole. Hum Mutat 30: E629–639, 2009. Heald B, Moran R , Milas M, Burke C, Eng C. Familial adenomatous polyposis in a patient with unexplained mental retardation. Nat Clin Pract Neurol 3: 694–700, 2007. Healy DL, Trounson AO, Andersen AN. Female infertility: causes and treatment. Lancet 343: 1539–1544, 1994. Heard PL, Carter EM, Crandall AC, Sebold C, Hale DE, Cody JD. High resolution genomic analysis of 18q- using oligo-microarray comparative genomic hybridization (aCGH). Am J Med Genet 149A : 1431–1437, 2009. Hecht F, Hecht BK . Linkage of skeletal dysplasia gene to t(2;8)(q32;p13) chromosome translocation breakpoint. Am J Med Genet 18: 779–780, 1984. Hegde MR , Fawkner M, Chong B, McGaughran J, Gilbert D, Love DR . Compound heterozygosity at the FMR1 gene. Genet Test 5: 135–138, 2001. Heidemann S, Plendl H, Vater I, et al. Maternal uniparental disomy 15 in a fetus resulting from a balanced familial translocation t(2;15) (p11;q11.2). Prenat Diagn 30: 183–185, 2010. Heilstedt HA , Ballif BC, Howard LA , et al. Physical map of 1p36, placement of breakpoints in monosomy 1p36, and clinical characterization of the syndrome. Am J Hum Genet 72: 1200–1212, 2003. Hellani A , Lauge A , Ozand P, Jaroudi K , Coskun S. Pregnancy after preimplantation genetic diagnosis for Ataxia Telangiectasia. Mol Hum Reprod 8: 785–788, 2002. Hempel M, Rivera Brugués N, Wagenstaller J, et al. Microdeletion syndrome 16p11.2-p12. 2: clinical and molecular characterization. Am J Med Genet 149A : 2106–2112, 2009. Hennebicq S, Pelletier R , Bergues U, Rousseaux S. Risk of trisomy 21 in offspring of patients with Klinefelter’s syndrome. Lancet 357: 2104–2105, 2001. Herens C, Jamar M, Alvarez-Gonzalez ML, et al. Private multiple congenital anomaly syndromes may result from unbalanced subtle translocations:
552
•
REFERENCES
t(2q;4p) explains the Lambotte syndrome. Am J Med Genet 73: 127–131, 1997. Herlihy AS, Halliday J. Is paternal age playing a role in the changing prevalence of Klinefelter syndrome? Eur J Hum Genet 16: 1173–1174, 2008. Herlihy AS, Halliday J, McLachlan R , Cock M, Gillam L. Assessing the risks and benefits of diagnosing genetic conditions with variable phenotypes through population screening: Klinefelter syndrome as an example. J Comm Genet 1: 41–46, 2010. Hernando C, Carrera M, Ribas I, et al. Prenatal and postnatal characterization of Y chromosome structural anomalies by molecular cytogenetic analysis. Prenat Diagn 22: 802–805, 2002. Heron SE, Scheffer IE, Grinton BE, et al. Familial neonatal seizures with intellectual disability caused by a microduplication of chromosome 2q24.3. Epilepsia 51: 1865–1869, 2010. Herrgård E, Mononen T, Mervaala E, et al. More severe epilepsy and cognitive impairment in the offspring of a mother with mosaicism for the ring 20 chromosome. Epilepsy Res 73: 122–128, 2007. Herzing LB, Cook EH, Ledbetter DH. Allele-specific expression analysis by RNA-FISH demonstrates preferential maternal expression of UBE3A and imprint maintenance within 15q11- q13 duplications. Hum Mol Genet 11: 1707–1718, 2002. Hewison J, Cuckle H, Baillie C, et al. Use of videotapes for viewing at home to inform choice in Down syndrome screening: a randomised controlled trial. Prenat Diagn 21: 146–149, 2001. Higgins AW, Alkuraya FS, Bosco AF, et al. Characterization of apparently balanced chromosomal rearrangements from the developmental genome anatomy project. Am J Hum Genet 82: 712–722, 2008. Hippman C, Oberlander TF, Honer WG, Misri S, Austin JC. Depression during pregnancy: the potential impact of increased risk for fetal aneuploidy on maternal mood. Clin Genet 75: 30–36, 2009. Hirsch B, Baldinger S. Pericentric inversion of chromosome 4 giving rise to dup(4p) and dup(4q) recombinants within a single kindred. Am J Med Genet 45: 5–8, 1993. Hitchins MP, Rickard S, Dhalla F, et al. Investigation of UBE3A and MECP2 in Angelman syndrome (AS) and patients with features of AS. Am J Med Genet 125A : 167–172, 2004. Hixon M, Millie E, Judis LA , et al. FISH studies of the sperm of fathers of paternally derived cases of trisomy 21: no evidence for an increase in aneuploidy. Hum Genet 103: 654–657, 1998.
Hochstenbach R , van Binsbergen E, Engelen J, et al. Array analysis and karyotyping: workflow consequences based on a retrospective study of 36,325 patients with idiopathic developmental delay in the Netherlands. Eur J Med Genet 52: 161–169, 2009. Hockey KA , Mulcahy MT, Montgomery P, Levitt S. Deletion of chromosome 5q and familial adenomatous polyposis. J Med Genet 26: 61–62, 1989. Höckner M, Utermann B, Erdel M, Fauth C, Utermann G, Kotzot D. Molecular characterization of a de novo ring chromosome 6 in a growth retarded but otherwise healthy woman. Am J Med Genet 146: 925–929, 2008. Hodge SE. Waiting for the amniocentesis. N Engl J Med 320: 63–64, 1989. Hodges CA , Ilagan A , Jennings D, Keri R , Nilson J, Hunt PA . Experimental evidence that changes in oocyte growth influence meiotic chromosome segregation. Hum Reprod 17: 1171–1180, 2002. Hodgson SV, Fagg NLK , Talbot IC, Wilkinson M. Deletions of the entire APC gene are associated with sessile colonic adenomas. J Med Genet 31: 426, 1994. Hogart A , Leung KN, Wang NJ, et al. Chromosome 15q11–13 duplication syndrome brain reveals epigenetic alterations in gene expression not predicted from copy number. J Med Genet 46: 86–93, 2009. Hogart A , Wu D, LaSalle JM, Schanen NC. The comorbidity of autism with the genomic disorders of chromosome 15q11.2-q13. Neurobiol Dis 38: 181–191, 2010. Höglund P, Holmberg C, de la Chapelle A , Kere J. Paternal isodisomy for chromosome 7 is compatible with normal growth and development in a patient with congenital chloride diarrhea. Am J Hum Genet 55: 747–752, 1994. Holden ST, Clarkson A , Thomas NS, Abbott K , James MR , Willatt L. A de novo duplication of Xp11.22-p11.4 in a girl with intellectual disability, structural brain anomalies, and preferential inactivation of the normal X chromosome. Am J Med Genet 152A : 1735–1740, 2010. Holinski-Feder E, Reyniers E, Uhrig S, et al. Familial mental retardation syndrome ATR-16 due to an inherited cryptic subtelomeric translocation, t(3;16)(q29;p13.3). Am J Hum Genet 66: 16–25, 2000. Holland CM. 47,XXX in an adolescent with premature ovarian failure and autoimmune disease. J Pediatr Adolesc Gynecol 14: 77–80, 2001. Hollox EJ, Armour JAL, Barber JCK . Extensive normal copy number variation of a beta-defensin
antimicrobial-gene cluster. Am J Hum Genet 73: 591–600, 2003. Hollox EJ, Atia T, Cross G, Parkin T, Armour JAL. High throughput screening of human subtelomeric DNA for copy number changes using multiplex amplifiable probe hybridisation (MAPH). J Med Genet 39: 790–795, 2002. Hollox EJ, Barber JCK , Brookes AJ, Armour JAL. Defensins and the dynamic genome: what we can learn from structural variation at human chromosome band 8p23.1. Genome Res 18: 1686–1697, 2008. Holmes-Siedle M, Ryynanen M, Lindenbaum RH. Parental decisions regarding termination of pregnancy following prenatal detection of sex chromosome abnormality. Prenat Diagn 7: 239–244, 1987. Homer L, Le Martelot MT, Morel F, et al. 45,X/46,XX mosaicism below 30% of aneuploidy: clinical implications in adult women from a reproductive medicine unit. Eur J Endocrinol 162: 617–623, 2010. Hoo JJ, Lorenz R , Fischer A , Fuhrmann W. Tiny interstitial duplication of proximal 7q in association with a maternal paracentric inversion. Hum Genet 62: 113–116, 1982. Hoo JJ, Lowry RB, Lin CC, Haslam RHA . Recurrent de novo interstitial deletion of 16q in two mentally retarded sisters. Clin Genet 27: 420–425, 1985. Hook EB. Exclusion of chromosomal mosaicism: tables of 90%, 95% and 99% confidence limits and comments on use. Am J Hum Genet 29: 94–97, 1977. Hook EB. Extra sex chromosomes and human behavior: the nature of the evidence regarding XYY, XXY, XXYY and XXX genotypes. In: Vallet HL, Porter IH, eds. Genetic Mechanisms of Sexual Development. New York: Academic Press, 1979: 437–463. Hook EB. Chromosome abnormalities and spontaneous fetal death following amniocentesis: further data and associations with maternal age. Am J Hum Genet 35: 110–116, 1983. Hook EB. Chromosome abnormalities: prevalence, risks and recurrence. In: Brock DJH, Rodeck CH, Ferguson-Smith MA, eds. Prenatal Diagnosis and Screening. Edinburgh, Scotland: Churchill Livingstone, 1992: 351–392. Hook EB, Cross PK . Interpretation of recent data pertinent to genetic counseling for Down syndrome: maternal-age-specific-rates, temporal trends, adjustments for paternal age, recurrence risks, risks after other cytogenetic abnormalities, recurrence risk after remarriage. In: Willey AM, Carter TP, Kelly S, Porter IH, eds. Clinical Genetics:
REFERENCES
•
553
Problems in Diagnosis and Counseling. New York: Academic Press, 1982: 119–139. Hook EB, Cross PK . Rates of mutant and inherited structural cytogenetic abnormalities detected at amniocentesis: results on about 63,000 fetuses. Ann Hum Genet 51: 27–55, 1987a. Hook EB, Cross PK . Extra structurally abnormal chromosomes (ESAC) detected at amniocentesis: frequency in approximately 75,000 prenatal cytogenetic diagnoses and associations with maternal and paternal age. Am J Hum Genet 40: 83–101, 1987b. Hopkin RJ, Schorry E, Bofinger M, et al. New insights into the phenotypes of 6q deletions. Am J Med Genet 70: 377–386, 1997. Hoppman-Chaney NL, Dawson DB, Nguyen L, et al. Partial hexasomy for the Prader-Willi-Angelman syndrome critical region due to a maternally inherited large supernumerary marker chromosome. Am J Med Genet 152A : 2034–2038, 2010. Horbinski C, Carter EM, Heard PL, et al. Molecular and clinical characterization of a recurrent cryptic unbalanced t(4q;18q) resulting in an 18q deletion and 4q duplication. Am J Med Genet 146A : 2898–2904, 2008. Horigome Y, Kondo I, Kuwajima K , Suzuki T. Familial occurrence of ring chromosome 15. Clin Genet 41: 178–180, 1992. Horike S, Ferreira JCP, Meguro-Horike M, et al. Screening of DNA methylation at the H19 promoter or the distal region of its ICR1 ensures efficient detection of chromosome 11p15 epimutations in Russell-Silver syndrome. Am J Med Genet 149A : 2415–2423, 2009. Horn D, Majewski F, Hildebrandt B, Körner H. Pallister-Killian syndrome: normal karyotype in prenatal chorionic villi, in postnatal lymphocytes, and in slowly growing epidermal cells, but mosaic tetrasomy 12p in skin fibroblasts. J Med Genet 32: 68–71, 1995. Horsley SW, Daniels RJ, Anguita E, et al. Monosomy for the most telomeric, gene-rich region of the short arm of human chromosome 16 causes minimal phenotypic effects. Eur J Hum Genet 9: 217–225, 2001. Horsman DE, Dill FJ, McGillivray BC, Kalousek DK . X chromosome aneuploidy in lymphocyte cultures from women with recurrent spontaneous abortions. Am J Med Genet 28: 981–987, 1987. Horsthemke B, Buiting K . Imprinting defects on human chromosome 15. Cytogenet Genome Res 113: 292–299, 2006. Horsthemke B, Maat-Kievit A , Sleegers E, et al. Familial translocations involving 15q11-q13 can
554
•
REFERENCES
give rise to interstitial deletions causing Prader-Willi or Angelman syndrome. J Med Genet 33: 848–851, 1996. Horsthemke B, Wagstaff J. Mechanisms of imprinting of the Prader-Willi/Angelman region. Am J Med Genet 146A : 2041–2052, 2008. Hoshi N, Fujita M, Mikuni M, et al. Seminoma in a postmenopausal woman with a Y;15 translocation in peripheral blood lymphocytes and a t(Y;15)/45,X Turner mosaic pattern in skin fibroblasts. J Med Genet 35: 852–856, 1998. Hosoki K , Takano K , Sudo A , Tanaka S, Saitoh S. Germline mosaicism of a novel UBE3A mutation in Angelman syndrome. Am J Med Genet 138A : 187–189, 2005. Hou JW, Tsai WY, Wang TR . Detection of KAL-1 gene deletion with fluorescence in situ hybridization. J Formos Med Assoc 98: 448–451, 1999. Hovatta O. Pregnancies in women with Turner’s syndrome. Ann Med 31: 106–110, 1999. Howarth ES, Konje JC, Healey KA , Duckett DP, Scudamore IW, Taylor DJ. Invasive testing for the karyotyping of mid-trimester intrauterine fetal death (IUFD): a pilot study. Prenat Diagn 22: 453–455, 2002. Howell RT, McDermott A , Gardner A , Dickinson V. Down’s syndrome with a recombinant tandem duplication of chromosome 21 derived from a maternal ring. J Med Genet 21: 310–314, 1984. Howell RT, Davies T. Diagnosis of Bloom’s syndrome by sister chromatid exchange evaluation in chorionic villus cultures. Prenat Diagn 14: 1071–1073, 1994. Hoyer J, Dreweke A , Becker C, et al. Molecular karyotyping in patients with mental retardation using 100K single-nucleotide polymorphism arrays. J Med Genet 44: 629–636, 2007. Hsu CC, Lee IW, Su MT, et al. Triple genetic identities for the complete hydatidiform mole, placenta and co-existing fetus after transfer of a single in vitro fertilized oocyte: case report and possible mechanisms. Hum Reprod 23: 2686–2691, 2008. Hsu LYF. Phenotype/karyotype correlations of Y chromosome aneuploidy with emphasis on structural aberrations in postnatally diagnosed cases. Am J Med Genet 53: 108–140, 1994. Hsu LYF. Prenatal diagnosis of 45,X/46,XX . Am J Hum Genet 58: 634–636, 1996. Hsu LYF, Benn PA . Revised guidelines for the diagnosis of mosaicism in amniocytes. Prenat Diagn 19: 1081–1082, 1999. Hsu LYF, Kaffe S, Jenkins EC, et al. Proposed guidelines for diagnosis of chromosome
mosaicism in amniocytes based on data derived from chromosome mosaicism and pseudomosaicism studies. Prenat Diagn 12: 555–573, 1992. Hsu LYF, Kaffe S, Perlis TE. A revisit of trisomy 20 mosaicism in prenatal diagnosis—an overview of 103 cases. Prenat Diagn 11: 7–15, 1991. Hsu LYF, Perlis TE. United States survey on chromosome mosaicism and pseudomosaicism in prenatal diagnosis. Prenat Diagn 4(spec no.): 97–130, 1984. Hsu LYF, Yu MT, Neu RL, et al. Rare trisomy mosaicism diagnosed in amniocytes, involving an autosome other than chromosomes 13, 18, 20, and 21: karyotype/phenotype correlations. Prenat Diagn 17: 201–242, 1997. Hsu LYF, Yu MT, Richkind KE, et al. Incidence and significance of chromosome mosaicism involving an autosomal structural abnormality diagnosed prenatally through amniocentesis: a collaborative study. Prenat Diagn 16: 1–28, 1996. Hsu WT, Shchepin DA , Mao R , et al. Mosaic trisomy 16 ascertained through amniocentesis: evaluation of 11 new cases. Am J Med Genet 80: 473–480, 1998. Huang B, Thangavelu M, Bhatt S, Sandlin C J, Wang S. Prenatal diagnosis of 45,X and 45,X mosaicism: the need for thorough cytogenetic and clinical evaluations. Prenat Diagn 22: 105–110, 2002. Hughes IA . Disorders of sex development: a new definition and classification. Best Pract Res Clin Endocrinol Metab 22: 119–134, 2008. Hughes IA , Houk C, Ahmed SF, Lee PA . Consensus statement on management of intersex disorders. Arch Dis Child 91: 554–563, 2006. Hulinsky R , Byrne JL, Lowichik A , Viskochil DH. Fetus with interstitial del(5)(p13.1p14.2) diagnosed postnatally with Cornelia de Lange syndrome. Am J Med Genet 137A : 336–338, 2005. Hultén M, Armstrong S, Challinor P, et al. Genomic imprinting in an Angelman and Prader-Willi translocation family. Lancet 338: 638–639, 1991. Hultén MA , Patel SD, Tankimanova M, et al. On the origin of trisomy 21 Down syndrome. Mol Cytogenet 1: 21, 2008. Hultén MA , Patel SD, Westgren M, et al. On the paternal origin of trisomy 21 Down syndrome. Mol Cytogenet 3: 4, 2010. Hundscheid RDL, Sistermans EA , Thomas CM, et al. Imprinting effect in premature ovarian failure confined to paternally inherited fragile X premutations. Am J Hum Genet 66: 413–418, 2000. Hung PH, Froenicke L, Lin CY, et al. Effects of environmental tobacco smoke in vivo on rhesus
monkey semen quality, sperm function, and sperm metabolism. Reprod Toxicol 27: 140–148, 2009. Hunt PA , Hassold TJ. Sex matters in meiosis. Science 296: 2181–2183, 2002. Hunter A , Brierley K , Tomkins D. 46,XX/46XY chromosome complement in amniotic fluid cell culture followed by the birth of a normal female child. Prenat Diagn 2: 127–131, 1982. Hunter AGW. Down syndrome. In: Cassidy SB, Allanson JE, eds. Management of Genetic Syndromes. New York: Wiley-Liss, 2001. Hunter AGW, Allanson JE. Follow-up study of patients with Wiedemann-Beckwith syndrome with emphasis on the change in facial appearance over time. Am J Med Genet 51: 102–107, 1994. Hunter JE, Epstein MP, Tinker SW, Charen KH, Sherman SL. Fragile X-associated primary ovarian insufficiency: evidence for additional genetic contributions to severity. Genet Epidemiol 32: 553–559, 2008. Hunter JE, Rohr JK , Sherman SL. Co-occurring diagnoses among FMR1 premutation allele carriers. Clin Genet 77: 374–381, 2010. Huret JL, Léonard C, Chery M, et al. Monosomy 21q: two cases of del(21q) and review of the literature. Clin Genet 48: 140–147, 1995. Hussain SZ, Evans AL, Ahmed OA , et al. Nonsyndromic mental retardation segregating with an apparently balanced t(1;17) reciprocal translocation through three generations. Am J Med Genet 95: 99–104, 2000. Hwang SH, Lee SM, Seo EJ, et al. A case of male infertility with a reciprocal translocation t(X;14) (p11.4;p12). Korean J Lab Med 27: 139–142, 2007. Hysert M, Bruyère H, Côté GB, et al. Prenatal cytogenetic assessment and inv(2)(p11.2q13). Prenat Diagn 26: 810–813, 2006. Iafrate AJ, Feuk L, Rivera MN, et al. Detection of large-scale variation in the human genome. Nat Genet 36: 949–951, 2004. Ichioka K , Utsunomiya N, Kohei N, Ueda N, Inoue K , Terai A . Adult onset of declining spermatogenesis in a man with nonmosaic Klinefelter’s syndrome. Fertil Steril 85: 1511 e1–2, 2006. Ideraabdullah FY, Vigneau S, Bartolomei MS. Genomic imprinting mechanisms in mammals. Mutat Res 647: 77–85, 2008. Ilberry PLT, Lee CWG, Winn SM. Incomplete trisomy in a mongoloid child exhibiting minimal stigmata. Med J Aust 48: 182–184, 1961. Iliopoulos D, Sekerli E, Vassiliou G, et al. Patau syndrome with a long survival (146 months): a clinical report and review of literature. Am J Med Genet 140: 92–93, 2006.
REFERENCES
•
555
Imai A , Takagi A , Tamaya T. A novel sex-determining region on Y (SRY) missense mutation identified in a 46,XY female and also in the father. Endocr J 46: 735–739, 1999. Ingelfinger FJ. Arrogance. N Engl J Med 303: 1507–1511, 1980. Inoue K , Dewar K , Katsanis N, et al. The 1.4-Mb CMT1A duplication/HNPP deletion genomic region reveals unique genome architectural features and provides insights into the recent evolution of new genes. Genome Res 11: 1018–1033, 2001. Irving MD, Buiting K , Kanber D, et al. Segmental paternal uniparental disomy (patUPD) of 14q32 with abnormal methylation elicits the characteristic features of complete patUPD14. Am J Med Genet 152A : 1942–1950, 2010. Irwin SA , Patel B, Idupulapati M, et al. Abnormal dendritic spine characteristics in the temporal and visual cortices of patients with fragile-X syndrome: a quantitative examination. Am J Med Genet 98: 161–167, 2001. Isaacs D. What is the value of a human baby? J Paediatr Child Health 38: 608–609, 2002. ISCN. An International System for Human Cytogenetic Nomenclature: Recommendations of the International Standing Committee on Human Cytogenetic Nomenclature. Shaffer LG, Slovak ML, Campbell LJ, eds. Basel, Switzerland: S. Karger, 2009. Iselius L, Lindsten J, Aurias A , et al. The 11q;22q translocation: a collaborative study of 20 new cases and analysis of 110 families. Hum Genet 64: 343–355, 1983. Ishii F, Fujita H, Nagai A , et al. Case report of rec(7) dup(7q)inv(7)(p22q22) and a review of the recombinants resulting from parental pericentric inversions on any chromosomes. Am J Med Genet 73: 290–295, 1997. Ishikawa S, Ishikawa M, Tokuda T, et al. Japanese family with an autosomal dominant chromosome instability syndrome: a new neurodegenerative disease? Am J Med Genet 94: 265–270, 2000. Ishikiriyama S, Tonoki H, Shibuya Y, et al. Waardenburg syndrome type I in a child with de novo inversion (2)(q35q37.3). Am J Med Genet 33: 505–507, 1989. Itoh N, Becroft DMO, Reeve AE, Morison IM. Proportion of cells with paternal 11p15 uniparental disomy correlates with organ enlargement in Wiedemann-beckwith syndrome. Am J Med Genet 92: 111–116, 2000. Itu M, Neelam T, Ammini AC, Kucheria K . Primary amenorrhoea in a triple X female. Aust NZ J Obstet Gynaecol 30: 386–388, 1990.
556
•
REFERENCES
Iwarsson E, Lundqvist M, Inzunza J, et al. A high degree of aneuploidy in frozen-thawed human preimplantation embryos. Hum Genet 104: 376–382, 1999. Iwarsson E, Malmgren H, Inzunza J, et al. Highly abnormal cleavage divisions in preimplantation embryos from translocation carriers. Prenat Diagn 20: 1038–1047, 2000. Iwarsson E, Sahlén S, Nordgren A . Jumping translocation in a phenotypically normal male: a study of mosaicism in spermatozoa, lymphocytes, and fibroblasts. Am J Med Genet 149A : 1706–1711, 2009. Izumi K , Culler D, Solomon BD, Muenke M, Parikh AS. Submicroscopic familial chromosomal translocation between 7q and 12p mimicking an autosomal dominant holoprosencephaly syndrome. Clin Genet 78: 402–404, 2010. Jaafar H, Gabriel-Robez O, Vignon F, Flori E, Rumpler Y. Supernumerary chromosomes and spermatogenesis in a human male carrier. Hum Genet 94: 74–76, 1994. Jaarola M, Martin RH, Ashley T. Direct evidence for suppression of recombination within two pericentric inversions in humans: a new spermFISH technique. Am J Hum Genet 63: 218–224, 1998. Jabs EW, Carpenter N. Molecular cytogenetic evidence for amplification of chromosome-specific alphoid sequences at enlarged C-bands on chromosome 6. Am J Hum Genet 43: 69–74, 1988. Jacobs P, Dalton P, James R , et al. Turner syndrome: a cytogenetic and molecular study. Ann Hum Genet 61: 471–483, 1997. Jacobs PA . Recurrence risks for chromosome abnormalities. Birth Defects: Orig Art Series 15:(5C) 71–80, 1979. Jacobs PA . Mutation rates of structural chromosome rearrangements in man. Am J Hum Genet 33: 44–54, 1981. Jacobs PA , Browne C, Gregson N, Joyce C, White H. Estimates of the frequency of chromosome abnormalities detectable in unselected newborns using moderate levels of banding. J Med Genet 29: 103–108, 1992. Jacobs PA , Strong JA . A case of human intersexuality having a possible XXY sex-determining mechanism. Nature 183: 302–303, 1959. Jacoby RF, Schlack S, Sekhon G, Laxova R . Del(10) (q22.3q24.1) associated with juvenile polyposis. Am J Med Genet 70: 361–364, 1997. Jacquemont S, Hagerman RJ, Hagerman PJ, Leehey MA . Fragile-X syndrome and fragile X-associated tremor/ataxia syndrome: two faces of FMR1. Lancet Neurol 6: 45–55, 2007.
Jacquemont S, Hagerman RJ, Leehey MA , et al. Penetrance of the fragile X-associated tremor/ ataxia syndrome in a premutation carrier population. J Am Med Assoc 291: 460–469, 2004. Jäger RJ, Harley VR , Pfeiffer RA , Goodfellow PN, Scherer G. A familial mutation in the testisdetermining gene SRY shared by both sexes. Hum Genet 90: 350–355, 1992. Jaillard S, Dubourg C, Gérard-Blanluet M, et al. 2q23.1 microdeletion identified by array comparative genomic hybridisation: an emerging phenotype with Angelman-like features? J Med Genet 46: 847–855, 2009. Jaju RJ, Fidler C, Haas OA , et al. A novel gene, NSD1, is fused to NUP98 in the t(5;11)(q35;p15.5) in de novo childhood acute myeloid leukemia. Blood 98: 1264–1267, 2001. Jalal SM, Kukolich MK , Garcia M, Day DW. Euchromatic 9q + heteromorphism in a family. Am J Med Genet 37: 155–156, 1990. Jalbert P, Jalbert H, Sele B. Types of imbalances in human reciprocal translocations: risks at birth. In: Daniel A, ed. The Cytogenetics of Mammalian Autosomal Rearrangements. New York: AR Liss, 1988. Jalbert P, Jalbert H, Sele B, et al. Partial trisomy for the long arms of chromosome no. 5 due to insertion and further “aneusomie de recombinaison.” J Med Genet 12: 418–423, 1975. Jalbert P, Sele B, Jalbert H. Reciprocal translocations: a way to predict the mode of imbalanced segregation by pachytene-diagram drawing. Hum Genet 55: 209–222, 1980. Jalil SSA , Mahran MA , Sule M. Placental mesenchymal dysplasia-can it be predicted prenatally? A case report. Prenat Diagn 29: 713–714, 2009. Jallinoja P, Santalahti P, Toiviainen H, Hemminki E. Acceptance of screening and abortion for Down syndrome among Finnish midwives and public health nurses. Prenat Diagn 19: 1015–1022, 1999. James PA , Gibson K , McGaughran J. Prenatal diagnosis of mosaic trisomy 20 in New Zealand. Aust NZ J Obstet Gynaecol 42: 486–489, 2002. James RS, Ellis K , Pettay D, Jacobs PA . Cytogenetic and molecular study of four couples with multiple trisomy 21 pregnancies. Eur J Hum Genet 6: 207–212, 1998. James RS, Sharp AJ, Cockwell AE, Coppin B, Jacobs PA . Evidence for a cryptic 46,XX cell line in a 45,X/46,X,psu idic(Xq) patient with normal reproduction. J Med Genet 34: 1030–1032, 1997. James RS, Temple IK , Dennis NR , Crolla JA . A search for uniparental disomy in carriers of
supernumerary marker chromosomes. Eur J Hum Genet 3: 21–26, 1995. James RS, Temple IK , Patch C, Thompson EM, Hassold T, Jacobs PA . A systematic search for uniparental disomy in carriers of chromosome translocations. Eur J Hum Genet 2: 83–95, 1994. Janke D. Centric fission of chromosome no. 7 in three generations. Hum Genet 60: 200–201, 1982. Jansen RPS, Bowman MC, de Boer KA , Leigh DA , Lieberman DB, McArthur SJ. What next for preimplantation genetic screening (PGS)? Experience with blastocyst biopsy and testing for aneuploidy. Hum Reprod 23: 1476–1478, 2008. Jaques AM, Collins VR , Muggli EE, et al. Uptake of prenatal diagnostic testing and the effectiveness of prenatal screening for Down syndrome. Prenat Diagn 30: 522–530, 2010. Jaques AM, Halliday JL, Bell RJ. Do women know that prenatal testing detects fetuses with Down syndrome? J Obstet Gynaecol 24: 647–651, 2004. Jaques AM, Halliday JL, Francis I, et al. Follow up and evaluation of the Victorian first-trimester combined screening programme for Down syndrome and trisomy 18. Br J Obstet Gynaecol 114: 812–818, 2007. Jarrah N, El-Shanti H, Khier A , Obeidat FN, Haddidi A , Ajlouni K . Familial disorder of sex determination in seven individuals from three related sibships. Eur J Pediatr 159: 912–918, 2000. Jarup L, Morris S, Richardson S, et al. Down syndrome in births near landfill sites. Prenat Diagn 27: 1191–1196, 2007. Jaruzelska J, Korcz A , Wojda A , et al. Mosaicism for 45,X cell line may accentuate the severity of spermatogenic defects in men with AZFc deletion. J Med Genet 38: 798–802, 2001. Jauch A , Robson L, Smith A . Investigations with fluorescence in situ hybridization (FISH) demonstrate loss of the telomeres on the reciprocal chromosome in three unbalanced translocations involving chromosome 15 in the Prader-Willi and Angelman syndromes. Hum Genet 96: 345–349, 1995. Jay A , Kilby MD, Roberts E, et al. Prenatal diagnosis of mosaicism for partial trisomy 8: a case report including fetal pathology. Prenat Diagn 19: 976–979, 1999. Jeffers MD, O’Dwyer P, Curran B, Leader M, Gillan JE. Partial hydatidiform mole: a common but underdiagnosed condition. A 3-year retrospective clinicopathological and DNA flow cytometric analysis. Int J Gynec Path 12: 315–323, 1993. Jenderny J, Caliebe A , Beyer C, Grote W. Transmission of a ring chromosome 18 from a mother with 46,XX/47,XX, + r(18) mosaicism to
REFERENCES
•
557
her daughter, resulting in a 46,XX,r(18) karyotype. J Med Genet 30: 964–965, 1993. Jenderny J, Jacobi ML, Rüger A , Röhrborn G. Chromosome aberrations in 450 sperm complements from eight controls and lack of increase after chemotherapy in two patients. Hum Genet 90: 151–154, 1992. Jenderny J, Schmidt W, Bartsch O. Inheritance of a t(13;14)(q10;q10) Robertsonian translocation with a low level of trisomy 13 mosaicism. Eur J Pediatr 169: 789–793, 2010. Jewell AF, Simpson GF, Pasztor L, Keene CL, Sullivan BA , Schwartz S. Prenatal diagnosis of two cases de novo dup(12p), identified by fluorescence in situ hybridization (FISH). Am J Hum Genet 51(suppl): 81A , 1992. Ji Y, Eichler EE, Schwartz S, Nicholls RD. Structure of chromosomal duplicons and their role in mediating human genomic disorders. Genome Res 10: 597–610, 2000. Jiang Y, Lucas I, Young DJ, et al. Common fragile sites are characterized by histone hypoacetylation. Hum Mol Genet 18: 4501–4512, 2009. Jiang YH, Martinez JE, Ou Z, et al. De novo and complex imbalanced chromosomal rearrangements revealed by array CGH in a patient with an abnormal phenotype and apparently “balanced” paracentric inversion of 14(q21q23). Am J Med Genet 146A : 1986–1993, 2008. Jinawath N, Zambrano R , Wohler E, et al. Mosaic trisomy 13: understanding origin using SNP array. J Med Genet 48: 323–326, 2011. Jobanputra V, Ash E, Anyane-Yeboa K , Warburton D, Levy B. Changes in an inherited ring (22) due to meiotic recombination? Implications for genetic counseling. Am J Med Genet 149A : 1310–1314, 2009. Jobanputra V, Chung WK , Hacker AM, Emanuel BS, Warburton D. A unique case of der(11)t(11;22),22 arising from 3:1 segregation of a maternal t(11;22) in a family with co-segregation of the translocation and breast cancer. Prenat Diagn 25: 683–686, 2005. Jobanputra V, Sobrino A , Kinney A , Kline J, Warburton D. Multiplex interphase FISH as a screen for common aneuploidies in spontaneous abortions. Hum Reprod 17: 1166–1170, 2002. Joenje H, Arwert F, Kwee ML, Madan K , Hoehn H. Confounding factors in the diagnosis of Fanconi anaemia. Am J Med Genet 79: 403–405, 1998. Johannisson R , Löhrs U, Wolff HH, Schwinger E. Two different XY-quadrivalent associations and impairment of fertility in men. Cytogenet Cell Genet 45: 222–230, 1987.
558
•
REFERENCES
Johannisson R , Winking H. Synaptonemal complexes of chains and rings in mice heterozygous for multiple Robertsonian translocations. Chromosome Res 2: 137–145, 1994. Johnson D, Horsley SW, Moloney DM, et al. A comprehensive screen for TWIST mutations in patients with craniosynostosis identifies a new microdeletion syndrome of chromosome band 7p21.1. Am J Hum Genet 63: 1282–1293, 1998. Johnson DS, Cinnioglu C, Ross R , et al. Comprehensive analysis of karyotypic mosaicism between trophectoderm and inner cell mass. Mol Hum Reprod 16(12): 944–949, 2010a. Johnson DS, Gemelos G, Baner J, et al. Preclinical validation of a microarray method for full molecular karyotyping of blastomeres in a 24-h protocol. Hum Reprod 25: 1066–1075, 2010b. Joly-Helas G, de La Rochebrochard C, MoussetSiméon N, et al. Complex chromosomal rearrangement and intracytoplasmic sperm injection: a case report. Hum Reprod 22: 1292–1297, 2007. Jones C, Booth C, Rita D, et al. Identification of a case of maternal uniparental disomy of chromosome 10 associated with confined placental mosaicism. Prenat Diagn 15: 843–848, 1995. Jones C, Slijepcevic P, Marsh S, et al. Physical linkage of the fragile site FRA11B and a Jacobsen syndrome chromosome deletion breakpoint in 11q23.3. Hum Mol Genet 3: 2123–2130, 1994. Jones HW. Problems of sex differentiation—surgical correction. Birth Defects Orig Artic Ser 14: 63–71, 1978. Jones HW, Veeck L. What is an embryo? Fertil Steril 77: 658–659, 2002. Joseph M, Cantú ES, Pai GS, Willi SM, Papenhausen PR , Weiss L. Xp pseudoautosomal gene haploinsufficiency and linear growth deficiency in three girls with chromosome Xp22; Yq11 translocation. J Med Genet 33: 906–911, 1996. Joseph-George AM, He Y, Marshall CR , et al. Euchromatic 9q13-q21 duplication variants are tandem segmental amplifications of sequence reciprocal to 9q13-q21 deletions. J Med Genet 48: 317–322, 2011. Jouannic JM, Tachdjian G, Costa JM, Bénifla JL. Coelomic fluid analysis: the absolute necessity to prove its fetal origin. Reprod Biomed Online 16: 148–151, 2008. Joyce CA , Dennis NR , Cooper S, Browne CE. Subtelomeric rearrangements: results from a study of selected and unselected probands with idiopathic mental retardation and control individuals by using high-resolution G-banding and FISH. Hum Genet 109: 440–451, 2001.
Joyce CA , Dennis NR , Howard F, Davis LM, Thomas NS. An 11p; 17p telomeric translocation in two families associated with recurrent miscarriages and Miller-Dieker syndrome. Eur J Hum Genet 10: 707–714, 2002. Jung E, Won HS, Lee PR , Kim A . Ultrasonographic measurement of fetal nasal bone length in the second trimester in Korean population. Prenat Diagn 27: 154–157, 2007. Kagami M, Nagai T, Fukami M, Yamazawa K , Ogata T. Silver-Russell syndrome in a girl born after in vitro fertilization: partial hypermethylation at the differentially methylated region of PEG1/MEST. J Assist Reprod Genet 24: 131–136, 2007. Kagami M, Nishimura G, Okuyama T, et al. Segmental and full paternal isodisomy for chromosome 14 in three patients: narrowing the critical region and implication for the clinical features. Am J Med Genet 138A : 127–132, 2005. Kagan KO, Anderson JM, Anwandter G, Neksasova K , Nicolaides KH. Screening for triploidy by the risk algorithms for trisomies 21, 18 and 13 at 11 weeks to 13 weeks and 6 days of gestation. Prenat Diagn 28: 1209–1213, 2008a. Kagan KO, Wright D, Valencia C, Maiz N, Nicolaides KH. Screening for trisomies 21, 18 and 13 by maternal age, fetal nuchal translucency, fetal heart rate, free beta-hCG and pregnancy-associated plasma protein-A . Hum Reprod 23: 1968–1975, 2008b. Kähkönen M, Kokkonen H-L, Haapala K , Winqvist R , Leisti J. Discrepancy in cytogenetic and DNA analyses of a patient with a deletion of proximal 15q due to familial pericentric inversion and analysis of eight other patients diagnosed to have Prader-Willi syndrome. Am J Hum Genet 47: A31, 1990. Kaiser P. Pericentric inversions. Problems and significance for clinical genetics. Hum Genet 68: 1–47, 1984. Kaiser P. Pericentric inversions: their problems and clinical significance. In: Daniel A, ed. The Cytogenetics of Mammalian Autosomal Rearrangements. New York: AR Liss, 1988: 163–247. Kajii T, Ferrier A , Niikawa N, Takahara H, Ohama K , Avirachan S. Anatomic and chromosomal anomalies in 639 spontaneous abortuses. Hum Genet 55: 87–98, 1980. Kallen B. Maternal use of oral contraceptives and Down syndrome. Contraception 39: 503–506, 1989. Kallen K . Down’s syndrome and maternal smoking in early pregnancy. Genet Epidemiol 14: 77–84, 1997.
Kalousek DK . Pathogenesis of chromosomal mosaicism and its effect on early human development. Am J Med Genet 91: 39–45, 2000. Kalousek DK , Langlois S, Robinson WP, et al. Trisomy 7 CVS mosaicism: pregnancy outcome, placental and DNA analysis in 14 cases. Am J Med Genet 65: 348–352, 1996. Kalscheuer VM, Musante L, Fang C, et al. A balanced chromosomal translocation disrupting ARHGEF9 is associated with epilepsy, anxiety, aggression, and mental retardation. Hum Mutat 30: 61–68, 2009. Kamiguchi Y, Rosenbusch B, Sterzik K , Mikamo K . Chromosomal analysis of unfertilized human oocytes prepared by a gradual fixation-air drying method. Hum Genet 90: 533–541, 1993. Kan AK , Abdalla HI, Oskarsson T. Two successful pregnancies in a 46,XY patient. Hum Reprod 12: 1434–1435, 1997. Kanaka-Gantenbein C, Papandreou E, Stefanaki K , et al. Spontaneous ovulation in a true hermaphrodite with normal male phenotype and a rare 46,XX/47,XXY Klinefelter’s mosaic karyotype. Horm Res 68: 139–144, 2007. Kanavakis E, Traeger-Synodinos J. Preimplantation genetic diagnosis in clinical practice. J Med Genet 39: 6–11, 2002. Kang SHL, Shaw C, Ou Z, et al. Insertional translocation detected using FISH confirmation of array-comparative genomic hybridization (aCGH) results. Am J Med Genet 152A : 1111–1126, 2010. Kantarci S, Donahoe PK . Congenital diaphragmatic hernia (CDH) etiology as revealed by pathway genetics. Am J Med Genet 145C: 217–226, 2007. Kara N, Okten G, Gunes SO, Saglam Y, Tasdemir HA , Pinarli FA . An epileptic case with mosaic ring chromosome 6 and 6q terminal deletion. Epilepsy Res 80: 219–223, 2008. Karadima G, Bugge M, Nicolaidis P, et al. Origin of nondisjunction in trisomy 8 and trisomy 8 mosaicism. Eur J Hum Genet 6: 432–438, 1998. Karaman B, Wollnik B, Ermis H, Yüksel-Apak M, Basaran S. A familial Xp+ chromosome detected during fetal karyotyping, which is associated with short stature in four generations of a Turkish family. Prenat Diagn 23: 336–339, 2003. Karanjawala ZE, Kääriäinen H, Ghosh S, et al. Complete maternal isodisomy of chromosome 8 in an individual with an early-onset ileal carcinoid tumor. Am J Med Genet 93: 207–210, 2000. Karatas JC, Barlow-Stewart K , Strong KA , Meiser B, McMahon C, Roberts C. Women’s experience of pre-implantation genetic diagnosis: a qualitative study. Prenat Diagn 30: 771–777, 2010. Kariminejad A , Kariminejad R , Moshtagh A , et al. Pericentric inversion of chromosome 18 in parents
REFERENCES
•
559
leading to a phenotypically normal child with segmental uniparental disomy 18. Eur J Hum Genet 19: 555–560, 2011. Karp LE. The terrible question. Am J Med Genet 14: 1–4, 1983. Kashork CD, Sutton VR , Fonda Allen JS, et al. Low or absent unconjugated estriol in pregnancy: an indicator for steroid sulfatase deficiency detectable by fluorescence in situ hybridization and biochemical analysis. Prenat Diagn 22: 1028–1032, 2002. Katari S, Turan N, Bibikova M, et al. DNA methylation and gene expression differences in children conceived in vitro or in vivo. Hum Mol Genet 18: 3769–3778, 2009. Kattera S, Chen C. Normal birth after microsurgical enucleation of tripronuclear human zygotes: case report. Hum Reprod 18: 1319–1322, 2003. Kaur A , Dhillon S, Garg PD, Singh JR . Ring chromosome 7 in an Indian woman. J Intellect Dev Disabil 33: 87–94, 2008. Kaur M, DeScipio C, McCallum J, et al. Precocious sister chromatid separation (PSCS) in Cornelia de Lange syndrome. Am J Med Genet 138: 27–31, 2005. Kausch K , Haaf T, Köhler J, Schmid M. Complex chromosomal rearrangement in a woman with multiple miscarriages. Am J Med Genet 31: 415–420, 1988. Kawano Y, Narahara H, Matsui N, Miyakawa I. Premature ovarian failure associated with a Robertsonian translocation. Acta Obstet Gynecol Scand 77: 467–469, 1998. Keagle MB, Gersen SL. Basic Laboratory Procedures. In: Gersen SL, Keagle MB, eds. The Principles of Clinical Cytogenetics. Totowa, NJ: Humana Press, 1999: 63–80. Keating DJ, Dubach D, Zanin MP, et al. DSCR1/ RCAN1 regulates vesicle exocytosis and fusion pore kinetics: implications for Down syndrome and Alzheimer’s disease. Hum Mol Genet 17: 1020–1030, 2008. Keeling SL, Lee-Jones L, Thompson P. Interstitial deletion 4q32–34 with ulnar deficiency: 4q33 may be the critical region in 4q terminal deletion syndrome. Am J Med Genet 99: 94–98, 2001. Kempe A , Engels H, Schubert R , et al. Familial ovarian dysgerminomas (Swyer syndrome) in females associated with 46 XY-karyotype. Gynecol Endocrinol 16: 107–111, 2002. Kennedy RD, D’Andrea AD. The Fanconi Anemia/ BRCA pathway: new faces in the crowd. Genes Dev 19: 2925–2940, 2005. Kennedy SJ, Teebi AS, Adatia I, Teshima I. Inherited duplication, dup (8) (p23.1p23.1) pat, in a father and daughter with congenital heart defects. Am J Med Genet 104: 79–80, 2001.
560
•
REFERENCES
Kennerknecht I, Barbi G, Vogel W. Maternal transmission of ring chromosome 21. Hum Genet 86: 99–101, 1990. Kennerknecht I, Just W, Vogel W. A triploid fetus with a diploid placenta: proposal of a mechanism. Prenat Diagn 13: 885–886, 1993a. Kennerknecht I, Vogel W, Mehnert K . A modified embryogenic model to explain embryonic/ extraembryonic chromosomal inconsistencies. Prenat Diagn 13: 1156–1159, 1993b. Kenney LB, Nicholson HS, Brasseux C, et al. Birth defects in offspring of adult survivors of childhood acute lymphoblastic leukemia. A Childrens Cancer Group/National Institutes of Health Report. Cancer 78: 169–176, 1996. Keppler-Noreuil KM, Carroll AJ, Finley SC, et al. Chromosome 18q paracentric inversion in a family with mental retardation and hearing loss. Am J Med Genet 76: 372–378, 1998. Kesler SR , Lightbody AA , Reiss AL. Cholinergic dysfunction in fragile X syndrome and potential intervention: a preliminary 1H MRS study. Am J Med Genet 149A : 403–407, 2009. Kessler S, Levine EK . Psychological aspects of genetic counseling. IV. The subjective assessment of probability. Am J Med Genet 28: 361–370, 1987. Keymolen K , Staessen C, Verpoest W, et al. A proposal for reproductive counselling in carriers of Robertsonian translocations: 10 years of experience with preimplantation genetic diagnosis. Hum Reprod 24: 2365–2371, 2009. Kher AS, Chattopadhyay A , Datta S, Kanade S, Sreenivasan VK , Bharucha BA . Familial mosaic Turner syndrome. Clin Genet 46: 382–383, 1994. Khoueiry R , Ibala-Rhomdane S, Méry L, et al. Dynamic CpG methylation of the KCNQ1OT1 gene during maturation of human oocytes. J Med Genet 45: 583–588, 2008. Kiholm Lund AB, Hove S, Kirchhoff M. A. 15q24 microduplication, reciprocal to the recently described 15q24 microdeletion, in a boy sharing clinical features with 15q24 microdeletion syndrome patients. Eur J Med Genet 51: 520–526, 2008. Kim ED, Bischoff FZ, Lipshultz LI, Lamb DJ. Genetic concerns for the subfertile male in the era of ICSI. Prenat Diagn 18: 1349–1365, 1998. Kim JH, Oh PS, Na HY, Kim SH, Cho HC. A case of mosaic ring chromosome 4 with subtelomeric 4p deletion. Korean J Lab Med 29: 77–81, 2009. Kim SR , Shaffer LG. Robertsonian translocations: mechanisms of formation, aneuploidy, and uniparental disomy and diagnostic considerations. Genet Test 6: 163–168, 2002. Kim Y, Lach FP, Desetty R , Hanenberg H, Auerbach AD, Smogorzewska A . Mutations of the SLX4
gene in Fanconi anemia. Nat Genet 43: 142–146, 2011. King PH, Waldrop R , Lupski JR , Shaffer LG. Charcot-Marie-Tooth phenotype produced by a duplicated PMP22 gene as part of a 17p trisomy-translocation to the X chromosome. Clin Genet 54: 413–416, 1998. Kingston HM, Nicolini U, Haslam J, Andrews T. 46,XY/47,XY, + 17p + mosaicism in amniocytes associated with fetal abnormalities despite normal fetal blood karyotype. Prenat Diagn 13: 637–642, 1993. Kircheisen R , Schroeder-Kurth T. Familiäres blasenmolen-syndrom und genetische aspekte dieser gestörten trophoblastentwicklung. Geburtshilfe Frauenheilkd 51: 569–571, 1991. Kirchhoff M, Rose H, Lundsteen C. High resolution comparative genomic hybridisation in clinical cytogenetics. J Med Genet 38: 740–744, 2001. Kirchhoff M, Bisgaard AM, Duno M, Hansen FJ, Schwartz M. A 17q21.31 microduplication, reciprocal to the newly described 17q21.31 microdeletion, in a girl with severe psychomotor developmental delay and dysmorphic craniofacial features. Eur J Med Genet 50: 256–263, 2007. Kirchhoff M, Bisgaard AM, Stoeva R , et al. Phenotype and 244k array-CGH characterization of chromosome 13q deletions: an update of the phenotypic map of 13q21.1-qter. Am J Med Genet 149A : 894–905, 2009. Kirchhoff M, Pedersen S, Kjeldsen E, et al. Prospective study comparing HR-CGH and subtelomeric FISH for investigation of individuals with mental retardation and dysmorphic features and an update of a study using only HR-CGH. Am J Med Genet 127A : 111–117, 2004. Kirchhoff M, Rose H, Lundsteen C. High resolution comparative genomic hybridisation in clinical cytogenetics. J Med Genet 38: 740–744, 2001. Kirkpatrick G, Ferguson KA , Gao H, et al. A comparison of sperm aneuploidy rates between infertile men with normal and abnormal karyotypes. Hum Reprod 23: 1679–1683, 2008. Kiss A , Rosa RFM, Dibi RP, et al. Chromosomal abnormalities in couples with history of recurrent abortion. Rev Bras Ginecol Obstet 31: 68–74, 2009. Kitsiou-Tzeli S, Manolakos E, Lagou M, et al. Characterization of a prenatally assessed de novo supernumerary minute ring chromosome 20 in a phenotypically normal male. Mol Cytogenet 2: 8, 2009. Kitsiou-Tzeli S, Sismani C, Ioannides M, et al. Array-CGH analysis and clinical description of 2q37.3 de novo subtelomeric deletion. Eur J Med Genet 50: 73–78, 2007.
Kitsiou-Tzeli S, Tzetis M, Sofocleous C, et al. De novo interstitial duplication of the 15q11.2-q14 PWS/ AS region of maternal origin: clinical description, array CGH analysis, and review of the literature. Am J Med Genet 152A : 1925–1932, 2010. Kivirikko S, Salonen R , Salo A, von Koskull H. Prenatally detected trisomy 7 mosaicism in a dysmorphic child. Prenat Diagn 22: 541–544, 2002. Kjær I, Niebuhr E. Studies of the cranial base in 23 patients with cri-du-chat syndrome suggest a cranial developmental field involved in the condition. Am J Med Genet 82: 6–14, 1999. Kleczkowska A , Fryns JP, Van den Berghe H. Pericentric inversions in man: personal experience and review of the literature. Hum Genet 75: 333–338, 1987. Kleczkowska A , Fryns JP, Vinken L, van den Berghe H. Effect of balanced X/autosome translocations on sexual and physical development. A personal experience in 4 patients. Clin Genet 27: 147–152, 1985. Kleefstra T, van Zelst-Stams WA, Nillesen WM, et al. Further clinical and molecular delineation of the 9q subtelomeric deletion syndrome supports a major contribution of EHMT1 haploinsufficiency to the core phenotype. J Med Genet 46: 598–606, 2009. Kleeman L, Bianchi DW, Shaffer LG, et al. Use of array comparative genomic hybridization for prenatal diagnosis of fetuses with sonographic anomalies and normal metaphase karyotype. Prenat Diagn 29: 1213–1217, 2009. Klein J, Graham JM, Platt LD, Schreck R . Trisomy 8 mosaicism in chorionic villus sampling: case report and counselling issues. Prenat Diagn 14: 451–454, 1994. Klein OD, Backstrand K , Cotter PD, Marco E, Sherr E, Slavotinek A . Case report: Y;6 translocation with deletion of 6p. Clin Dysmorphol 14: 93–96, 2005. Kligman I, Benadiva C, Alikani M, Munné S. The presence of multinucleated blastomeres in human embryos is correlated with chromosomal abnormalities. Hum Reprod 11: 1492–1498, 1996. Klinger K , Landes G, Shook D, et al. Rapid detection of chromosome aneuploidies in uncultured amniocytes by using fluorescence in situ hybridization (FISH). Am J Hum Genet 51: 55–65, 1992. Klopocki E, Schulze H, Strauss G, et al. Complex inheritance pattern resembling autosomal recessive inheritance involving a microdeletion in thrombocytopenia-absent radius syndrome. Am J Hum Genet 80: 232–240, 2007. Knegt AC, Li S, Engelen JJM, Bijlsma EK , Warburton PE. Prenatal diagnosis of a karyotypically normal
REFERENCES
•
561
pregnancy in a mother with a supernumerary neocentric 13q21→13q22 chromosome and balancing reciprocal deletion. Prenat Diagn 23: 215–220, 2003. Knight MA , Hernandez D, Diede SJ, et al. A duplication at chromosome 11q12.2–11q12.3 is associated with spinocerebellar ataxia type 20. Hum Mol Genet 17: 3847–3853, 2008. Knight SJL, Flint J. Perfect endings: a review of subtelomeric probes and their use in clinical diagnosis. J Med Genet 37: 401–409, 2000. Knoppers BM. “Well-bear and well-rear” in China? Am J Hum Genet 63: 686–687, 1998. Ko DS, Cho JW, Park SY, et al. Clinical outcomes of preimplantation genetic diagnosis (PGD) and analysis of meiotic segregation modes in reciprocal translocation carriers. Am J Med Genet 152A : 1428–1433, 2010. Ko PC, Peng HH, Soong YK , Chang SD. Triplet pregnancy complicated with one hydatidiform mole and preeclampsia in a 46,XY female with gonadal dysgenesis. Taiwan J Obstet Gynecol 46: 276–280, 2007. Kobayashi T, Narahara K , Yokoyama Y, et al. Gardner syndrome in a boy with interstitial deletion of the long arm of chromosome 5. Am J Med Genet 41: 460–463, 1991. Koç A, Arisoy O, Pala E, et al. Prenatal diagnosis of mosaic ring 22 duplication/deletion with terminal 22q13 deletion due to abnormal first trimester screening and choroid plexus cyst detected on ultrasound. J Obstet Gynaecol Res 35: 978–982, 2009. Koç A , Kan D, Karaer K , et al. An unexpected finding in a child with neurological problems: mosaic ring chromosome 18. Eur J Pediatr 167: 655–659, 2008. Koeberl DD, McGillivray B, Sybert VP. Prenatal diagnosis of 45,X/46,XX mosaicism and 45,X: implications for postnatal outcome. Am J Hum Genet 57: 661–666, 1995. Kojis TL, Gatti RA , Sparkes RS. The cytogenetics of ataxia telangiectasia. Cancer Genet Cytogenet 56: 143–156, 1991. Kokalj Vokac N, Seme Ciglenecki P, Erjavec A , Zagradisnik B, Zagorac A . Partial Xp duplication in a girl with dysmorphic features: the change in replication pattern of late-replicating dupX chromosome. Clin Genet 61: 54–61, 2002. Kokkonen H, Leisti J. An unexpected recurrence of Angelman syndrome suggestive of maternal germ-line mosaicism of del(15)(q11q13) in a Finnish family. Hum Genet 107: 83–85, 2000. Komori S, Nakata Y, Sakata K , Kato H, Koyoma K . Analysis for microdeletions of Y chromosome in a single spermatozoon from a man with severe oligozoospermia. J Hum Genet 46: 76–79, 2001.
562
•
REFERENCES
Kompanje EJO. The earliest description of an autopsy on a case of Roberts syndrome reported in 1672: some additions. Am J Med Genet 149A : 1610–1611, 2009. Kondo Y, Mizuno S, Ohara K , et al. Two cases of partial trisomy 21 (pter-q22.1) without the major features of Down syndrome. Am J Med Genet 140: 227–232, 2006. Kondoh Y, Toma T, Ohashi H, et al. Inv dup del(4) (:p14→p16. 3::p16.3→qter) with manifestations of partial duplication 4p and Wolf-Hirschhorn syndrome. Am J Med Genet 120A : 123–126, 2003. Konialis C, Hagnefelt B, Sevastidou S, et al. Uncovering recurrent microdeletion syndromes and subtelomeric deletions/duplications through non-selective application of a MLPA-based extended prenatal panel in routine prenatal diagnosis. Prenat Diagn 31: 571–577, 2011. Koochek M, Harvard C, Hildebrand MJ, et al. 15q duplication associated with autism in a multiplex family with a familial cryptic translocation t(14;15)(q11.2;q13.3) detected using array-CGH. Clin Genet 69: 124–134, 2006. Kooij L, Tymstra T, Berg P. The attitude of women toward current and future possibilities of diagnostic testing in maternal blood using fetal DNA . Prenat Diagn 29: 164–168, 2009. Kooper AJA , Faas BHW, Kater-Baats E, et al. Multiplex ligation-dependent probe amplification (MLPA) as a stand-alone test for rapid aneuploidy detection in amniotic fluid cells. Prenat Diagn 28: 1004–1010, 2008. Koorey D, Basha NJ, Tomaras C, Freiman J, Robson L, Smith A . Appendiceal carcinoma complicating adenomatous polyposis in a young woman with a de novo constitutional reciprocal translocation t(5;8)(q22; p23.1). J Med Genet 37: 71–75, 2000. Korenberg JR , Chen XN, Schipper R , et al. Down syndrome phenotypes: the consequences of chromosomal imbalance. Proc Natl Acad Sci USA 91: 4997–5001, 1994. Kosaki R , Migita O, Takahashi T, Kosaki K . Two distinctive classic genetic syndromes, 22q11.2 deletion syndrome and Angelman syndrome, occurring within the same family. Am J Med Genet 149A : 702–705, 2009. Kosho T, Sakazume S, Kawame H, et al. De-novo balanced translocation between 7q31 and 10p14 in a girl with central precocious puberty, moderate mental retardation, and severe speech impairment. Clin Dysmorphol 17: 31–34, 2008. Koskinen S, Onnelainen T, de la Chapelle A, Kere J. A rare reciprocal translocation (12;21) segregating for nine generations. Hum Genet 92: 509–512, 1993.
Koster MPH, Pennings JLA , Imholz S, et al. Proteomics and Down syndrome screening: a validation study. Prenat Diagn 30: 1039–1043, 2010. Kosztolányi G. Does “ring syndrome” exist? An analysis of 207 case reports on patients with a ring autosome. Hum Genet 75: 174–179, 1987. Kosztolányi G, Brecevic L, Bajnòczky K , Schinzel A , Riegel M. Mosaic supernumerary ring chromosome 1 in a three-generational family: 10-year follow-up report. Eur J Med Genet 54: 152–156, 2011. Kosztolányi G, Méhes K , Hook EB. Inherited ring chromosomes: an analysis of published cases. Hum Genet 87: 320–324, 1991. Kotzot D. Abnormal phenotypes in uniparental disomy (UPD): fundamental aspects and a critical review with bibliography of UPD other than 15. Am J Med Genet 82: 265–274, 1999. Kotzot D. Complex and segmental uniparental disomy (UPD): review and lessons from rare chromosomal complements. J Med Genet 38: 497–507, 2001. Kotzot D. Review and meta-analysis of systematic searches for uniparental disomy (UPD) other than UPD 15. Am J Med Genet 111: 366–375, 2002. Kotzot D. Complex and segmental uniparental disomy updated. J Med Genet 45: 545–556, 2008a. Kotzot D. Prenatal testing for uniparental disomy: indications and clinical relevance. Ultrasound Obstet Gynecol 31: 100–105, 2008b. Kotzot D, Hoffmann K , Kujat A , Holland H, Froster UG, Mücke J. Implications of FISH investigations in MIDAS syndrome associated with a 46,XX,t(X;Y) karyotype. Am J Med Genet 113: 108–110, 2002. Kotzot D, Holland H, Keller E, Froster UG. Maternal isochromosome 7q and paternal isochromosome 7p in a boy with growth retardation. Am J Med Genet 102: 169–172, 2001. Kotzot D, Lurie IW, Mehes K , Werder E, Schinzel A . No evidence of uniparental disomy 2, 6, 14, 16, 20, and 22 as a major cause of intrauterine growth retardation. Clin Genet 58: 177–180, 2000a. Kotzot D, Martinez MJ, Bagci G, et al. Parental origin and mechanisms of formation of cytogenetically recognisable de novo direct and inverted duplications. J Med Genet 37: 281–286, 2000b. Kovaleva NV. Germ-line transmission of trisomy 21: Data from 80 families suggest an implication of grandmaternal age and a high frequency of female-specific trisomy rescue. Mol Cytogenet 3: 7, 2010. Kovaleva NV, Shaffer LG. Under-ascertainment of mosaic carriers of balanced homologous
acrocentric translocations and isochromosomes. Am J Med Genet 121A : 180–187, 2003. Kozma C, Slavotinek AM, Meck JM. Segregation of a t(1;3) translocation in multiple affected family members with both types of adjacent-1 segregants. Am J Med Genet 124A : 118–128, 2004. Kraff J, Tang HT, Cilia R , et al. Screen for excess FMR1 premutation alleles among males with parkinsonism. Arch Neurol 64: 1002–1006, 2007. Kramer RL, Jarve RK , Yaron Y, et al. Determinants of parental decisions after the prenatal diagnosis of Down syndrome. Am J Med Genet 79: 172–174, 1998. Krantz ID, McCallum J, DeScipio C, et al. Cornelia de Lange syndrome is caused by mutations in NIPBL, the human homolog of Drosophila melanogaster Nipped-B. Nat Genet 36: 631–635, 2004. Krantz ID, Rand EB, Genin A , et al. Deletions of 20p12 in Alagille syndrome: frequency and molecular characterization. Am J Med Genet 70: 80–86, 1997. Krausz C, McElreavey K . Y chromosome microdeletions in “fertile” males. Hum Reprod 16: 1306–1307, 2001. Krieg SA , Lathi RB, Behr B, Westphal LM. Normal pregnancy after tetraploid karyotype on trophectoderm biopsy. Fertil Steril 92: 1169 e10, 2009. Krob G, Braun A , Kuhnle U. True hermaphroditism: geographical distribution, clinical findings, chromosomes and gonadal histology. Eur J Pediatr 153: 2–10, 1994. Kroepfl T, Petek E, Schwarzbraun T, Kroisel PM, Plecko B. Mental retardation in a girl with a subtelomeric deletion on chromosome 20q and complete deletion of the myelin transcription factor 1 gene (MYT1). Clin Genet 73: 492–495, 2008. Kroisel PM, Petek E, Wagner K . Phenotype of five patients with Greig syndrome and microdeletion of 7p13. Am J Med Genet 102: 243–249, 2001. Krüger G, Gótz J, Dunker H, Pelz L. Isochromosome (18q) in siblings. Clin Genet 32: 249–253, 1987. Krzeminska D, Steinfeld C, Cloez JL, et al. Prenatal diagnosis of Williams syndrome based on ultrasound signs. Prenat Diagn 29: 710–712, 2009. Kubota T, Nonoyama S, Tonoki H, et al. A new assay for the analysis of X-chromosome inactivation based on methylation-specific PCR . Hum Genet 104: 49–55, 1999. Kuchinka BD, Barrett IJ, Moya G, et al. Two cases of confined placental mosaicism for chromosome 4, including one with maternal uniparental disomy. Prenat Diagn 21: 36–39, 2001. Kuechler A , Hauffa BP, Köninger A , et al. An unbalanced translocation unmasks a recessive
REFERENCES
•
563
mutation in the follicle-stimulating hormone receptor (FSHR) gene and causes FSH resistance. Eur J Hum Genet 18: 656–661, 2010. Kühl H, Röttger S, Heilbronner H, Enders H, Schempp W. Loss of the Y chromosomal PAR2-region in four familial cases of satellited Y chromosomes (Yqs). Chromosome Res 9: 215–222, 2001. Kulharya AS, Mills Lovell C, Flannery DB. Unusual mosaic karyotype resulting from adjacent 1 segregation of t(11;22): importance of performing skin fibroblast karyotype in patients with unexplained multiple congenital anomalies. Am J Med Genet 113: 367–370, 2002. Kulharya AS, Roop H, Kukolich MK , Nachtman RG, Belmont JW, Garcia-Heras J. Mild phenotypic effects of a de novo deletion Xpter→Xp22.3 and duplication 3pter→3p23. Am J Med Genet 56: 16–21, 1995. Kuller JA , Meyer WR , Traynor KD, Hartmann KE. Disposition of sperm donors with resultant abnormal pregnancies. Hum Reprod 16: 1553–1555, 2001. Kumar A , Cassidy SB, Romero L, Schwartz S. Molecular cytogenetics of a de novo interstitial deletion of chromosome arm 6q in a developmentally normal girl. Am J Med Genet 86: 227–231, 1999. Kumar RA , KaraMohamed S, Sudi J, et al. Recurrent 16p11.2 microdeletions in autism. Hum Mol Genet 17: 628–638, 2008. Kumra S, Wiggs E, Krasnewich D, et al. Brief report: association of sex chromosome anomalies with childhood-onset psychotic disorders. J Am Acad Child Adolesc Psychiatry 37: 292–296, 1998. Kunz J, Schoner K , Stein W, Rehder H, Fritz B. Tetrasomy 12p (Pallister-Killian syndrome): difficulties in prenatal diagnosis. Arch Gynecol Obstet 280: 1049–1053, 2009. Kuo PL. Maternal trisomy 21 mosaicism and recurrent spontaneous abortion. Fertil Steril 78: 432–433, 2002. Kupchik GS, Barrett SK , Babu A , Charria-Ortiz G, Velinov M, Macera MJ. Atypical 18p- syndrome associated with partial trisomy 16p in a chromosomally unbalanced child of consanguineous parents with an identical balanced translocation. Eur J Med Genet 48: 57–65, 2005. Kuppermann M, Learman LA , Gates E, et al. Beyond race or ethnicity and socioeconomic status: predictors of prenatal testing for Down syndrome. Obstet Gynecol 107: 1087–1097, 2006. Kurahashi H, Emanuel BS. Unexpectedly high rate of de novo constitutional t(11;22) translocations in
564
•
REFERENCES
sperm from normal males. Nat Genet 29: 139–140, 2001. Kurahashi H, Sakamoto M, Ono J, Honda A , Okada S, Nakamura Y. Molecular cloning of the chromosomal breakpoint in the LIS1 gene of a patient with isolated lissencephaly and balanced t(8;17). Hum Genet 103: 189–192, 1998. Kurahashi H, Inagaki H, Ohye T, et al. The constitutional t(11;22): implications for a novel mechanism responsible for gross chromosomal rearrangements. Clin Genet 78: 299–309, 2010. Kuroda-Kawaguchi T, Skaletsky H, Brown LG, et al. The AZFc region of the Y chromosome features massive palindromes and uniform recurrent deletions in infertile men. Nat Genet 29: 279–286, 2001. Kurotaki N, Imaizumi K , Harada N, et al. Haploinsufficiency of NSD1 causes Sotos syndrome. Nat Genet 30: 365–366, 2002. Kurz MW, Schlitter AM, Klenk Y, et al. FMR1 alleles in Parkinson’s disease: relation to cognitive decline and hallucinations, a longitudinal study. J Geriatr Psychiatry Neurol 20: 89–92, 2007. Kvaløy K , Galvagni F, Brown WRA . The sequence organization of the long arm pseudoautosomal region of the human sex chromosomes. Hum Mol Genet 3: 771–778, 1994. Lacassie Y, Arriaza MI, Vargas A , La Motta I. Ring 2 chromosome: ten-year follow-up report. Am J Med Genet 85: 117–122, 1999. Lacbawan FL, White BJ, Anguiano A , et al. Rare interstitial deletion (2)(p11.2p13) in a child with pericentric inversion (2)(p11.2q13) of paternal origin. Am J Med Genet 87: 139–142, 1999. Lachlan KL, Youings S, Costa T, Jacobs PA , Thomas NS. A clinical and molecular study of 26 females with Xp deletions with special emphasis on inherited deletions. Hum Genet 118: 640–651, 2006. Lacombe D, Saura R , Taine L, Battin J. Confirmation of assignment of a locus for Rubinstein-Taybi syndrome gene to 16p13.3. Am J Med Genet 44: 126–128, 1992. Lafay-Cousin L, Payne E, Strother D, Chernos J, Chan M, Bernier FP. Goldenhar phenotype in a child with distal 22q11.2 deletion and intracranial atypical teratoid rhabdoid tumor. Am J Med Genet 149A : 2855–2859, 2009. Lagier-Tourenne C, Ginglinger E, Alembik Y, et al. Two cousins with partial trisomy 12q and monosomy 12p recombinants of a familial pericentric inversion of the chromosome 12. Am J Med Genet 125A : 77–85, 2004. Lahn BT, Ma N, Breg WR , Stratton R , Surti U, Page DC. Xq-Yq interchange resulting in
supernormal X-linked gene expression in severely retarded males with 46,XYq- karyotype. Nat Genet 8: 243–250, 1994. Lalani SR , Thakuria JV, Cox GF, et al. 20p12.3 microdeletion predisposes to Wolff-ParkinsonWhite syndrome with variable neurocognitive deficits. J Med Genet 46: 168–175, 2009. Lam YH, Lee CP, Sin SY, et al. Comparison and integration of first trimester fetal nuchal translucency and second trimester maternal serum screening for fetal Down syndrome. Prenat Diagn 22: 730–735, 2002. Lambert-Messerlian G, Dugoff L, Vidaver J, et al. First- and second-trimester Down syndrome screening markers in pregnancies achieved through assisted reproductive technologies (ART): a FASTER trial study. Prenat Diagn 26: 672–678, 2006. Lanasa MC, Hogge WA . X chromosome defects as an etiology of recurrent spontaneous abortion. Semin Reprod Med 18: 97–103, 2000. Lancet Editorial. Western eyes on China’s eugenics law. Lancet 346: 131, 1995. Landwehr C, Montag M, van der Ven K , Weber RG. Rapid comparative genomic hybridization protocol for prenatal diagnosis and its application to aneuploidy screening of human polar bodies. Fertil Steril 90: 488–496, 2008. Langlois S, Yong PJ, Yong SL, et al. Postnatal follow-up of prenatally diagnosed trisomy 16 mosaicism. Prenat Diagn 26: 548–558, 2006. Larsen SO, Hansen J, Pedersen BN. Expected, prenatally discovered, and born cases of Down syndrome in Denmark during the period 1980–1998. Prenat Diagn 21: 630–633, 2001. Lathi RB, Mark SD, Westphal LM, Milki AA . Cytogenetic testing of anembryonic pregnancies compared to embryonic missed abortions. J Assist Reprod Genet 24: 521–524, 2007. Lathi RB, Westphal LM, Milki AA . Aneuploidy in the miscarriages of infertile women and the potential benefit of preimplanation genetic diagnosis. Fertil Steril 89: 353–357, 2008. Lau NM, Huang JY, MacDonald S, et al. Feasibility of fertility preservation in young females with Turner syndrome. Reprod Biomed Online 18: 290–295, 2009. Lau TK , Fung HYM, Rogers MS, Cheung KL. Racial variation in incidence of trisomy 21: survey of 57,742 Chinese deliveries. Am J Med Genet 75: 386–388, 1998. Laufer-Cahana A , Krantz ID, Bason LD, Lu FM, Piccoli DA , Spinner NB. Alagille syndrome inherited from a phenotypically normal mother with a mosaic 20p microdeletion. Am J Med Genet 112: 190–193, 2002.
Laureys G, Speleman F, Opdenakker G, Benoit Y, Leroy J. Constitutional translocation t(1;17)(p36; q12–21) in a patient with neuroblastoma. Genes Chromosomes Cancer 2: 252–254, 1990. Lautrup CK , Kjaergaard S, Brøndum-Nielsen K , et al. Testing for 22q11 microdeletion in 146 fetuses with nuchal translucency above the 99th percentile and a normal karyotype. Acta Obstet Gynecol Scand 87: 1252–1255, 2008. Lavery SA , Aurell R , Turner C, et al. Preimplantation genetic diagnosis: patients’ experiences and attitudes. Hum Reprod 17: 2464–2467, 2002. Layman LC. Human gene mutations causing infertility. J Med Genet 39: 153–161, 2002. Layman LC, Tho SPT, Clark AD, Kulharya A , McDonough PG. Phenotypic spectrum of 45,X/46,XY males with a ring Y chromosome and bilaterally descended testes. Fertil Steril 91: 791–797, 2009. Lazzaro SJ, Speevak MD, Farrell SA . Recombinant Down syndrome: a case report and literature review. Clin Genet 59: 128–130, 2001. Le Caignec C, Boceno M, Saugier-Veber P, et al. Detection of genomic imbalances by array based comparative genomic hybridisation in fetuses with multiple malformations. J Med Genet 42: 121–128, 2005. Leach NT, Chudoba I, Stewart TV, Holmes LB, Weremowicz S. Maternally inherited duplication of chromosome 7, dup(7)(p11.2p12), associated with mild cognitive deficit without features of Silver-Russell syndrome. Am J Med Genet 143A : 1489–1493, 2007. Leach NT, Cole SM, Sandstrom DJ, Weremowicz S. A novel pericentric inversion of chromosome 14 involving the rRNA gene cluster. Prenat Diagn 25: 620–621, 2005. Lebo RV Prenatal diagnosis of Charcot-Marie-Tooth disease. Prenat Diagn 18: 169–172, 1998. Lebo RV, Milunsky J, Higgins AW, Loose B, Huang XL, Wyandt HE. Symmetric replication of an unstable isodicentric Xq chromosome derived from isolocal maternal sister chromatid recombination. Am J Med Genet 85: 429–437, 1999. Lebo RV, Shapiro LR , Fenerci EY, et al. Rare etiology of autosomal recessive disease in a child with noncarrier parents. Am J Hum Genet 67: 750–754, 2000. Lebre AS, Morinière V, Dunand O, Bensman A , Morichon-Delvallez N, Antignac C. Maternal uniparental heterodisomy of chromosome 17 in a patient with nephropathic cystinosis. Eur J Hum Genet 17: 1019–1023, 2009.
REFERENCES
•
565
Lecce R , Murdolo M, Gelli G, et al. The euchromatic 9p+ polymorphism is a locus-specific amplification caused by repeated copies of a small DNA segment mapping within 9p12. Hum Genet 118: 760–766, 2006. Leclercq G, Buvat-Herbaut M, Monnier JC, Vinatier D, Dufour P. Turner’s syndrome and pregnancy in donor oocytes and in vitro fertilization. Three case reports. J Gynecol Obstet Biol Reprod (Paris) 21: 635–640, 1992. Leclercq S, Baron X , Jacquemont ML, Cuillier F, Cartault F. Mosaic trisomy 22: five new cases with variable outcomes. Implications for genetic counselling and clinical management. Prenat Diagn 30: 168–172, 2010. Ledbetter DH, Rich DC, O’Connell P, Leppert M, Carey JC. Precise localization of NF1 to 17q11.2 by balanced translocation. Am J Hum Genet 44: 20–24, 1989. Ledbetter DH, Zachary JM, Simpson JL, et al. Cytogenetic results from the U.S. Collaborative Study on CVS. Prenat Diagn 12: 317–345, 1992. Lee IW, Su MT, Hsu CC, Lin YH, Chen PY, Kuo PL. Constitutional complex chromosomal rearrangements in azoospermic men—case report and literature review. Urology 68:1343.e5–8, 2006. Lee J, Stanley JR , Vaz SA , et al. Down syndrome with pure partial trisomy 21q22 due to a paternal insertion (4;21) uncovered by uncultured amniotic fluid interphase FISH. Am J Med Genet 132A : 206–208, 2005. Lee JA , Carvalho CM, Lupski JR . A DNA replication mechanism for generating nonrecurrent rearrangements associated with genomic disorders. Cell 131: 1235–1247, 2007. Lee M, Munné S. Pregnancy after polar body biopsy and freezing and thawing of human embryos. Fertil Steril 73: 645–647, 2000. Lee MH, Park SY, Kim YM, et al. Prenatal diagnosis of a familial complex chromosomal rearrangement involving chromosomes 5, 10, 16 and 18. Prenat Diagn 22: 102–104, 2002. Leegte B, Sikkema-Raddatz B, Hordijk R , et al. Three cases of mosaicism for balanced reciprocal translocations. Am J Med Genet 79: 362–365, 1998. Lefort G, Blanchet P, Belgrade N, et al. Stable dicentric duplication-deficiency chromosome 14 resulting from crossing-over within a maternal paracentric inversion. Am J Med Genet 113: 333–338, 2002. Lefort G, Blanchet P, Chaze AM, et al. Cytogenetic and molecular study of a jumping translocation in a baby with Dandy-Walker malformation. J Med Genet 38: 67–73, 2001.
566
•
REFERENCES
Légaré F, St-Jacques S, Gagnon S, et al. Prenatal screening for Down syndrome: a survey of willingness in women and family physicians to engage in shared decision-making. Prenat Diagn 31: 319–326, 2011. Leggett V, Jacobs P, Nation K , Scerif G, Bishop DV. Neurocognitive outcomes of individuals with a sex chromosome trisomy: XXX, XYY, or XXY: a systematic review. Dev Med Child Neurol 52: 119–129, 2010. Leichtman DA , Schmickel RD, Gelehrter TD, Judd WJ, Woodbury MC, Meilinger KL. Familial Turner syndrome. Ann Intern Med 89: 473–476, 1978. Leisti JT, Kaback MM, Rimoin DL. Human X-autosome translocations: differential inactivation of the X chromosome in a kindred with an X-9 translocation. Am J Hum Genet 27: 441–453, 1975. Lejeune J. Types et contretypes. J Génét Hum 15(suppl): 20–32, 1966. Lejeune J. The William Allan Memorial Award Lecture: On the nature of men. Am J Hum Genet 22: 121–128, 1970. Lejeune J, Gautier M, Turpin R . Étude des chromosomes somatiques de neuf enfants mongoliens. Comp Rend Hebd Séances Acad Sci 248: 1721–1722, 1959. Lejeune J, Lafourcade J, Berger R , et al. Trois cas de délétion partielle du bras court d’un chromosome 5. Comp Rend Hebd Séances Acad Sci 257: 3098–3102, 1963. Lemyre E, der Kaloustian VM, Duncan AMV. Stable non-Robertsonian dicentric chromosomes: four new cases and a review. J Med Genet 38: 76–79, 2001. Lenzi ML, Smith J, Snowden T, et al. Extreme heterogeneity in the molecular events leading to the establishment of chiasmata during meiosis i in human oocytes. Am J Hum Genet 76: 112–127, 2005. Leonard NJ, Tomkins DJ. Diploid/tetraploid/t(1;6) mosaicism in a 17-year-old female with hypomelanosis of Ito, multiple congenital anomalies, and body asymmetry. Am J Med Genet 112: 86–90, 2002. Leonard S, Bower C, Petterson B, Leonard H. Survival of infants born with Down’s syndrome: 1980–96. Paediatr Perinat Epidemiol 14: 163–171, 2000. Lespinasse J, Gicquel C, Robert M, Le Bouc Y. Phenotypic and genotypic variability in monozygotic triplets with Turner syndrome. Clin Genet 54: 56–59, 1998. Lespinasse J, Hoffmann P, Lauge A , et al. Chromosomal instability in two siblings with
gonad deficiency: case report. Hum Reprod 20: 158–162, 2005. Lestou VS, Desilets V, Lomax BL, et al. Comparative genomic hybridization: a new approach to screening for intrauterine complete or mosaic aneuploidy. Am J Med Genet 92: 281–284, 2000. Letterie GS. Unique unbalanced X;X translocation (Xq22;p11.2) in a woman with primary amenorrhea but without Ullrich-Turner syndrome. Am J Med Genet 59: 414–416, 1995. Levitas AS, Reid CS. An angel with Down syndrome in a sixteenth century Flemish Nativity painting. Am J Med Genet 116A : 399–405, 2003. Levy B, Dunn TM, Hirschhorn K , Kardon N. Jumping translocations in spontaneous abortions. Cytogenet Cell Genet 88: 25–29, 2000. Levy MJ, Stillman RJ. Reproductive potential in survivors of childhood malignancy. Pediatrician 18: 61–70, 1991. Levy-Mozziconacci A , Piquet C, Scheiner C, et al. i(18q) in amniotic and fetal cells with a normal karyotype in direct chorionic villus sampling: cytogenetics and pathology. Prenat Diagn 16: 1156–1159, 1996. Lewi L, Blickstein I, Van Schoubroeck D, et al. Diagnosis and management of heterokaryotypic monochorionic twins. Am J Med Genet 140: 272–275, 2006. Lewin B. Genes V. New York: Oxford University Press, 1994. Lewis-Jones I, Aziz N, Seshadri S, Douglas A , Howard P. Sperm chromosomal abnormalities are linked to sperm morphologic deformities. Fertil Steril 79: 212–215, 2003. Li F, Lisi EC, Wohler ES, Hamosh A , Batista DA . 3q29 interstitial microdeletion syndrome: an inherited case associated with cardiac defect and normal cognition. Eur J Med Genet 52: 349–352, 2009. Li M, Squire JA , Weksberg R . Molecular genetics of Wiedemann-Beckwith syndrome. Am J Med Genet 79: 253–259, 1998. Li S, Hassed S, Mulvihill JJ, Nair AK , Hopcus DJ. Double trisomy. Am J Med Genet 124A : 96–98, 2004. Liao C, Li DZ. Pregnancy outcome following prenatal diagnosis of sex chromosome abnormalities in mainland China. Prenat Diagn 28: 443–444, 2008. Lichtenbelt KD, Hochstenbach R , van Dam WM, Eleveld MJ, Poot M, Beemer FA . Supernumerary ring chromosome 7 mosaicism: case report, investigation of the gene content, and delineation of the phenotype. Am J Med Genet 132A : 93–100, 2005. Liebaers I, Desmyttere S, Verpoest W, et al. Report on a consecutive series of 581 children born after
blastomere biopsy for preimplantation genetic diagnosis. Hum Reprod 25: 275–282, 2010. Liehr T. Characterization of prenatally assessed de novo small supernumerary marker chromosomes by molecular cytogenetics. Methods Mol Biol 444: 27–38, 2008. Liehr T. Cytogenetic contribution to uniparental disomy (UPD). Mol Cytogenet 3: 8, 2010. Liehr T, Claussen U, Starke H. Small supernumerary marker chromosomes (sSMC) in humans. Cytogenet Genome Res 107: 55–67, 2004. Liehr T, Mrasek K , Weise A , et al. Small supernumerary marker chromosomes—progress towards a genotype-phenotype correlation. Cytogenet Genome Res 112: 23–34, 2006. Lim AST, Lim TH, Kee SK , Chieng R , Tay SK . Sperm segregation patterns by fluorescence in situ hybridization studies of a 46,XY,t(2;6) heterozygote giving rise to a rare triploid product of conception with a 69,XXY,t(2;6)(p12;q24) der(6)t(2;6)(p12;q24)pat karyotype. Am J Med Genet 117A : 172–176, 2003. Lim AST, Su LC. Mosaic trisomy 18 male with normal intelligence who fathered a normal baby girl. Am J Med Genet 76: 365–366, 1998. Lim CK , Cho JW, Kim JY, Kang IS, Shim SH, Jun JH. A healthy live birth after successful preimplantation genetic diagnosis for carriers of complex chromosome rearrangements. Fertil Steril 90: 1680–1684, 2008a. Lim CK , Cho JW, Song IO, Kang IS, Yoon YD, Jun JH. Estimation of chromosomal imbalances in preimplantation embryos from preimplantation genetic diagnosis cycles of reciprocal translocations with or without acrocentric chromosomes. Fertil Steril 90:2144–2151, 2008b. Lim D, Bowdin SC, Tee L, et al. Clinical and molecular genetic features of BeckwithWiedemann syndrome associated with assisted reproductive technologies. Hum Reprod 24: 741–747, 2009. Lim HJ, Kim YJ, Yang JH, et al. Amniotic fluid interphase fluorescence in situ hybridization (FISH) for detection of aneuploidy; experiences in 130 prenatal cases. J Korean Med Sci 17: 589–592, 2002. Lin CC, Hsieh YY, Wang CH, et al. Prenatal detection and characterization of a small supernumerary marker chromosome (sSMC) derived from chromosome 22 with apparently normal phenotype. Prenat Diagn 26: 898–902, 2006. Lin YH, Chuang L, Lin YM, Teng YN, Kuo PL. Isochromosome of Yp in a man with Sertoli-cellonly syndrome. Fertil Steril 83: 764–766, 2005.
REFERENCES
•
567
Linardopoulou EV, Williams EM, Fan Y, Friedman C, Young JM, Trask BJ. Human subtelomeres are hot spots of interchromosomal recombination and segmental duplication. Nature 437: 94–100, 2005. Lindblom A , Sandelin K , Iselius L, et al. Predisposition for breast cancer in carriers of constitutional translocation 11q;22q. Am J Hum Genet 54: 871–876, 1994. Linden MG, Bender BG. Fifty-one prenatally diagnosed children and adolescents with sex chromosome abnormalities. Am J Med Genet 110: 11–18, 2002. Linden MG, Bender BG. Reply to correspondence from Haverty et al.—“47,XXX associated with malformations.” Am J Med Genet 125A : 112, 2004. Linden MG, Bender BG, Robinson A . Sex chromosome tetrasomy and pentasomy. Pediatrics 96: 672–682, 1995. Linden MG, Bender BG, Robinson A . Genetic counseling for sex chromosome abnormalities. Am J Med Genet 110: 3–10, 2002. Linden MG, Tassone F, Gane LW, Hills JL, Hagerman RJ, Taylor AK . Compound heterozygous female with fragile X syndrome. Am J Med Genet 83: 318–321, 1999. Lindenbaum RH, Hultén M, McDermott A , Seabright M. The prevalence of translocations in parents of children with regular trisomy 21: a possible interchromosomal effect? J Med Genet 22: 24–28, 1985. Liou JD, Chen CP, Breg WR , Hobbins JC, Mahoney MJ, Yang-Feng TL. Fetal blood sampling and cytogenetic abnormalities. Prenat Diagn 13: 1–8, 1993. Liou JD, Ma YY, Gibson LH, et al. Cytogenetic and molecular studies of a familial paracentric inversion of Y chromosome present in a patient with ambiguous genitalia. Am J Med Genet 70: 134–137, 1997. Lippe B. Turner syndrome. Endocrinol Metab Clin North Am 20: 121–152, 1991. Lisi EC, Hamosh A , Doheny KF, et al. 3q29 interstitial microduplication: a new syndrome in a threegeneration family. Am J Med Genet 146A : 601–609, 2008. Lissoni S, Baronchelli S, Villa N, et al. Chromosome territories, X;Y translocation and premature ovarian failure: is there a relationship? Mol Cytogenet 2: 19, 2009. Little J. The Chernobyl accident, congenital anomalies and other reproductive outcomes. Paediatr Perinat Epidemiol 7: 121–151, 1993. Loeffler J, Soelder E, Erdel M, et al. Muellerian aplasia associated with ring chromosome 8p12q12 mosaicism. Am J Med Genet 116A : 290–294, 2003.
568
•
REFERENCES
Loesch DZ, Bui QM, Huggins RM, Mitchell RJ, Hagerman RJ, Tassone F. Transcript levels of the intermediate size or grey zone fragile X mental retardation 1 alleles are raised, and correlate with the number of CGG repeats. J Med Genet 44: 200–204, 2007. Loesch DZ, Cook M, Litewka L, et al. A low symptomatic form of neurodegeneration in younger carriers of the FMR1 premutation, manifesting typical radiological changes. J Med Genet 45: 179–181, 2008. Loesch DZ, Huggins R , Hay DA , Gedeon AK , Mulley JC, Sutherland GR . Genotype-phenotype relationships in fragile X syndrome: a family study. Am J Hum Genet 53: 1064–1073, 1993. Loesch DZ, Khaniani MS, Slater HR , et al. Small CGG repeat expansion alleles of FMR1 gene are associated with parkinsonism. Clin Genet 76: 471–476, 2009. Löffler J, Utermann B, Duba HC, Mayr U, Utermann G, Erdel M. Mental and psychomotoric retardation in two brothers with pure partial trisomy 7q32q34 due to a maternal insertion (14;7). Am J Med Genet 91: 291–297, 2000. Lonardo F, Cantalupo G, Ciavarella M, et al. Prenatal diagnosis of 46,XX testicular DSD. Molecular, cytogenetic, molecular-cytogenetic, and ultrasonographic evaluation. Prenat Diagn 29: 998–1001, 2009. Lopes J, Ravisé N, Vandenberghe A , et al. Fine mapping of de novo CMT1A and HNPP rearrangements within CMT1A-REPs evidences two distinct sex-dependent mechanisms and candidate sequences involved in recombination. Hum Mol Genet 7: 141–148, 1998. López Andreu JA , Fernández PJ, Ferrís i Tortajada J, et al. Persistent altered spermatogenesis in long-term childhood cancer survivors. Pediatr Hematol Oncol 17: 21–30, 2000. López-Expósito I, Bafalliu JA , Santos M, Fuster C, Puche-Mira A , Guillen-Navarro E. Intrachromosomal partial triplication of chromosome 13 secondary to a paternal duplication with mild phenotypic effect. Am J Med Genet 146A : 1190–1194, 2008. López-Pajares I, Delicado A , Lapunzina P, et al. Tetrasomy 8p: discordance of amniotic fluid and blood karyotypes. Am J Med Genet 118A : 353–357, 2003. López Pajares I, Villa O, Salido M, et al. Euchromatic variant 16p+. Implications in prenatal diagnosis. Prenat Diagn 26: 535–538, 2006. Lorda-Sánchez I, Diego-Alvarez D, Ayuso C, de Alba MR , Trujillo MJ, Ramos C. Trisomy 2 due to a 3:1
segregation in an abortion studied by QF-PCR and CGH. Prenat Diagn 25: 934–938, 2005. Lorenz P, Rupprecht E, Tellkamp H. An unusual type of acrocephalosyndactyly with bilateral parietooccipital “encephalocele,” micropenis, and severe mental retardation. Am J Med Genet 36: 265–268, 1990. Losekoot M, Hoogendoorn E, Olmer R , et al. Prenatal diagnosis of the fragile X syndrome: loss of mutation owing to a double recombinant or gene conversion event at the FMR1 locus. J Med Genet 34: 924–926, 1997. Lowe X , Eskenazi B, Nelson DO, Kidd S, Alme A , Wyrobek AJ. Frequency of XY sperm increases with age in fathers of boys with Klinefelter syndrome. Am J Hum Genet 69: 1046–1054, 2001. Lowry RB, Sibbald B, Bedard T. Alberta congenital anomalies surveillance system: eighth report 1980–2007. Edmonton: Alberta Health and Wellness, 2009. Lu X , Shaw CA , Patel A , et al. Clinical implementation of chromosomal microarray analysis: summary of 2513 postnatal cases. PloS One 2:e327, 2007. Lubinsky MS. Kouska’s fallacy: the error of the divided denominator. Lancet 2: 1449–1450, 1986. Lubs HA . A marker X chromosome. Am J Hum Genet 21: 231–244, 1969. Luciani JM, Guichaoua MR , Delafontaine D, North MO, Gabriel-Robez O, Rumpler Y. Pachytene analysis in a 17;21 reciprocal translocation carrier: role of the acrocentric chromosomes in male sterility. Hum Genet 77: 246–250, 1987. Luciani JJ, de Mas P, Depetris D, et al. Telomeric 22q13 deletions resulting from rings, simple deletions, and translocations: cytogenetic, molecular, and clinical analyses of 32 new observations. J Med Genet 40: 690–696, 2003. Lüdecke HJ, Schmidt O, Nardmann J, et al. Genes and chromosomal breakpoints in the Langer-Giedion syndrome region on human chromosome 8. Hum Genet 105: 619–628, 1999. Luetjens CM, Rolf C, Gassner P, Werny JE, Nieschlag E. Sperm aneuploidy rates in younger and older men. Hum Reprod 17: 1826–1832, 2002. Luijsterburg AJM, van der Zee DC, Gaillard JLJ, et al. Chorionic villus sampling and end-artery disruption of the fetus. Prenat Diagn 17: 71–76, 1997. Lukusa T, Devriendt K , Holvoet M, Fryns JP. Dicentric chromosome 9 due to tandem duplication of the 9p11-q13 region: unusual chromosome 9 variant. Am J Med Genet 91: 192–197, 2000. Lukusa T, Fryns JP, Kleczkowska A , Van den Berghe H. Role of gonadal dysgenesis in gonadoblastoma
induction in 46, XY individuals. The Leuven experience in 46, XY pure gonadal dysgenesis and testicular feminization syndromes. Genet Couns 2: 9–16, 1991. Lun FM, Jin YY, Sun H, et al. Noninvasive prenatal diagnosis of a case of Down syndrome due to Robertsonian translocation by massively parallel sequencing of maternal plasma DNA . Clin Chem 57: 917–919, 2011. Lundin J, Söderhäll C, Lundén L, et al. 22q11.2 microduplication in two patients with bladder exstrophy and hearing impairment. Eur J Med Genet 53: 61–65, 2010. Lungeanu A , Arghir A , Arps S, et al. Chromosome Y isodicentrics in two cases with ambiguous genitalia and features of Turner syndrome. Balkan J Med Genet 11: 8, 2008. Lupski JR . Genomic disorders ten years on. Genome Med 1: 42, 2009. Lybæk H, Øyen N, Fauske L, Houge G. A 2.1 Mb deletion adjacent but distal to a 14q21q23 paracentric inversion in a family with spherocytosis and severe learning difficulties. Clin Genet 74: 553–559, 2008. Lyle R , Prandini P, Osoegawa K , et al. Islands of euchromatin-like sequence and expressed polymorphic sequences within the short arm of human chromosome 21. Genome Res 17: 1690–1696, 2007. Ma S, Yuen BH, Penaherrera M, Koehn D, Ness L, Robinson W. ICSI and the transmission of X-autosomal translocation: a three-generation evaluation of X;20 translocation: case report. Hum Reprod 18: 1377–1382, 2003. Maas NMC, Van Buggenhout G, Hannes F, et al. Genotype-phenotype correlation in 21 patients with Wolf-Hirschhorn syndrome using high resolution array comparative genome hybridisation (CGH). J Med Genet 45: 71–80, 2008. Maas NMC, Van Vooren S, Hannes F, et al. The t(4;8) is mediated by homologous recombination between olfactory receptor gene clusters, but other 4p16 translocations occur at random. Genet Couns 18: 357–365, 2007. Maas NMC, Vermeesch JR , Fryns JP. A male with two idic(Y)(q12) chromosomes: a distinct phenotype resembling the XXXY/XXXXY syndrome. Am J Med Genet 138A : 294–296, 2005. Macayran JF, Cederbaum SD, Fox MA . Diagnostic yield of chromosome analysis in patients with developmental delay or mental retardation who are otherwise nondysmorphic. Am J Med Genet 140: 2320–2323, 2006.
REFERENCES
•
569
MacDermot KD, Jack E, Cooke A , et al. Investigation of three patients with the “ring syndrome,” including familial transmission of ring 5, and estimation of reproductive risks. Hum Genet 85: 516–520, 1990. MacDonald M, Hassold T, Harvey J, Wang LH, Morton NE, Jacobs P. The origin of 47,XXY and 47,XXX aneuploidy: heterogeneous mechanisms and role of aberrant recombination. Hum Mol Genet 3: 1365–1371, 1994. Machev N, Gosset P, Warter S, Treger M, Schillinger M, Viville S. Fluorescence in situ hybridization sperm analysis of six translocation carriers provides evidence of an interchromosomal effect. Fertil Steril 84: 365–373, 2005. Macintosh MCM. Perception of risk. In: Grudzinskas JG, Chard T, Chapman M, Cuckle H, eds. Screening for Down’s Syndrome. Cambridge, England: Cambridge University Press, 1994: 19–23. Mackay DJG, Boonen SE, Clayton-Smith J, et al. A maternal hypomethylation syndrome presenting as transient neonatal diabetes mellitus. Hum Genet 120: 262–269, 2006. Mackie Ogilvie C, Harrison RH, Horsley SW, Hodgson SV, Kearney L. A mitotically stable marker chromosome negative for whole chromosome libraries, centromere probes and chromosome specific telomere regions: a novel class of supernumerary marker chromosome? Cytogenet Cell Genet 92: 69–73, 2001. Mackie Ogilvie C, Scriven PN. Meiotic outcomes in reciprocal translocation carriers ascertained in 3-day human embryos. Eur J Hum Genet 10: 801–806, 2002. Mackie Ogilvie C, Yaron Y, Beaudet AL. Current controversies in prenatal diagnosis 3: For prenatal diagnosis, should we offer less or more than metaphase karyotyping? Prenat Diagn 29: 11–14, 2009. Madan K . Paracentric inversions: a review. Hum Genet 96: 503–515, 1995. Madan K , Hompes PGA , Schoemaker J, Ford CE. X-autosome translocation with a breakpoint in Xq22 in a fertile woman and her 47,XXX infertile daughter. Hum Genet 59: 290–296, 1981. Madan K , Kleinhout J. First trimester abortions associated with a translocation t(1;20)(p36;p11). Hum Genet 76: 109, 1987. Madan K , Menko FH. Intrachromosomal insertions: a case report and a review. Hum Genet 89: 1–9, 1992. Madan K , Nieuwint AWM. Reproductive risks for paracentric inversion heterozygotes: inversion or insertion? That is the question. Am J Med Genet 107: 340–343, 2002.
570
•
REFERENCES
Madan K , Nieuwint AWM, van Bever Y. Recombination in a balanced complex translocation of a mother leading to a balanced reciprocal translocation in the child. Review of 60 cases of balanced complex translocations. Hum Genet 99: 806–815, 1997. Madariaga ML, Rivera H. Familial inv(X) (p22q22): ovarian dysgenesis in two sisters with del Xq and fertility in one male carrier. Clin Genet 52: 180–183, 1997. Mademont-Soler I, Morales C, Madrigal I, et al. Prenatal diagnosis of two different unbalanced forms of an inherited (Y;12) translocation. Am J Med Genet 149A : 2820–2823, 2009. Magee AC, Humphreys MW, McKee S, Stewart M, Nevin NC. De novo direct duplication 2 (p12→p21) with paternally inherited pericentric inversion 2p11.2 2q12.2. Clin Genet 54: 65–69, 1998a. Magee AC, Nevin NC, Armstrong MJ, McGibbon D, Nevin J. Ullrich-Turner syndrome: seven pregnancies in an apparent 45,X woman. Am J Med Genet 75: 1–3, 1998b. Magenis E, Webb MJ, Spears B, Opitz JM. Blaschkolinear malformation syndrome in complex trisomy-7 mosaicism. Am J Med Genet 87: 375–383, 1999. Mahmood R , Brierley CH, Faed MJW, Mills JA , Delhanty JDA . Mechanisms of maternal aneuploidy: FISH analysis of oocytes and polar bodies in patients undergoing assisted conception. Hum Genet 106: 620–626, 2000. Makrakis E, Papagianni V, Deligeoroglou E. A unique case of trichorionic quadruplet pregnancy after oocyte donation in a patient with Turner’s syndrome. Gynecol Endocrinol 25: 460–463, 2009. Makrydimas G, Sebire NJ, Thornton SE, Zagorianakou N, Lolis D, Fisher RA . Complete hydatidiform mole and normal live birth: a novel case of confined placental mosaicism: case report. Hum Reprod 17: 2459–2463, 2002. Makrydimas G, Sotiriadis A , Spencer K , Cowans NJ, Nicolaides KH. ADAM12-s in coelomic fluid and maternal serum in early pregnancy. Prenat Diagn 26: 1197–1200, 2006. Malan V, De Blois MC, Prieur M, et al. Sotos syndrome caused by a paracentric inversion disrupting the NSD1 gene. Clin Genet 73: 89–91, 2008. Malan V, Gesny R , Morichon-Delvallez N, et al. Prenatal diagnosis and normal outcome of a 46,XX/46,XY chimera: a case report. Hum Reprod 22: 1037–1041, 2007. Malan V, Raoul O, Firth HV, et al. 19q13.11 deletion syndrome: a novel clinically recognisable genetic
condition identified by array comparative genomic hybridisation. J Med Genet 46: 635–640, 2009. Malvestiti F, De Toffol S, Chinetti S, et al. Prenatal diagnosis of del(4)(q27q31.23), due to a maternal balanced complex chromosome rearrangement, characterized by array-CGH. Prenat Diagn 30:280–283, 2010. Malzac P, Webber H, Moncla A , et al. Mutation analysis of UBE3A in Angelman syndrome patients. Am J Hum Genet 62: 1353–1360, 1998. Mange A , Desmetz C, Bellet V, Molinari N, Maudelonde T, Solassol J. Proteomic profile determination of autosomal aneuploidies by mass spectrometry on amniotic fluids. Proteome Sci 6: 1, 2008. Manipalviratn S, DeCherney A , Segars J. Imprinting disorders and assisted reproductive technology. Fertil Steril 91: 305–315, 2009. Mann NP, Fitzsimmons J, Fitzsimmons E, Cooke P. Roberts syndrome: clinical and cytogenetic aspects. J Med Genet 19: 116–119, 1982. Manning M, Hudgins L; Professional Practice and Guidelines Committee. Array-based technology and recommendations for utilization in medical genetics practice for detection of chromosomal abnormalities. Genet Med 12: 742–745, 2010. Manolakos E, Vetro A , Kitmirides S, et al. Prenatal diagnosis of a fetus with ring chromosome 15 characterized by array-CGH. Prenat Diagn 29: 884–888, 2009. Mantzouratou A , Mania A , Apergi M, Laver S, Serhal P, Delhanty J. Meiotic and mitotic behaviour of a ring/deleted chromosome 22 in human embryos determined by preimplantation genetic diagnosis for a maternal carrier. Mol Cytogenet 2: 3, 2009. Maraia R , Saal HM, Wangsa D. A chromosome 17q de novo paracentric inversion in a patient with campomelic dysplasia; case report and etiologic hypothesis. Clin Genet 39: 401–408, 1991. Maranda B, Lemieux N, Lemyre E. Familial deletion 18p syndrome: case report. BMC Med Genet 7: 60, 2006. Marangi G, Leuzzi V, Orteschi D, et al. Duplication of the Rubinstein-Taybi region on 16p13.3 is associated with a distinctive phenotype. Am J Med Genet 146A : 2313–2317, 2008. Maraschio P, Tupler R , Dainotti E, Cortinovis M, Tiepolo L. Molecular analysis of a human Y;1 translocation in an azoospermic male. Cytogenet Cell Genet 65: 256–260, 1994. Marchetti F, Bishop JB, Lowe X , Generoso WM, Hozier J, Wyrobek AJ. Etoposide induces heritable chromosomal aberrations and aneuploidy during male meiosis in the mouse. Proc Natl Acad Sci USA 98: 3952–3957, 2001.
Margarit E, Coll MD, Oliva R , Gómez D, Soler A , Ballesta F. SRY gene transferred to the long arm of the X chromosome in a Y-positive XX true hermaphrodite. Am J Med Genet 90: 25–28, 2000. Margarit E, Soler A , Carrió A , et al. Molecular, cytogenetic, and clinical characterisation of six XX males including one prenatal diagnosis. J Med Genet 35: 727–730, 1998. Marinescu RC, Johnson EI, Dykens EM, Hodapp RM, Overhauser J. No relationship between the size of the deletion and the level of developmental delay in cri-du-chat syndrome. Am J Med Genet 86: 66–70, 1999a. Marinescu RC, Mainardi PC, Collins MR , et al. Growth charts for cri-du-chat syndrome: an international collaborative study. Am J Med Genet 94: 153–162, 2000. Marinescu RC, Mamunes P, Kline AD, Schmidt J, Rojas K , Overhauser J. Variability in a family with an insertion involving 5p. Am J Med Genet 86: 258–263, 1999b. Marion JP, Fernhoff PM, Korotkin J, Priest JH. Preand postnatal diagnosis of trisomy 4 mosaicism. Am J Med Genet 37: 362–365, 1990. Mark HFL. Medical Cytogenetics. New York: Marcel Dekker, 2000. Mark HFL, Mendoza T, Abuelo D, Beauregard LJ, May JB, LaMarche PH. Reproduction in a woman with low percentage t(21q21q) mosaicism. J Med Genet 14: 221–223, 1977. Markkanen A , Ruutu T, Rasi V, Franssila K , Knuutila S, de la Chapelle A . Constitutional translocation t(3;6)(p14;p11) in a family with hematologic malignancies. Cancer Genet Cytogenet 25: 87–95, 1987. Marmor D, Duyck F. Male reproductive potential after MOPP therapy for Hodgkin’s disease: a long-term survey. Andrologia 27: 99–106, 1995. Maron JL, Bianchi DW. Prenatal diagnosis using cell-free nucleic acids in maternal body fluids: a decade of progress. Am J Med Genet 145C: 5–17, 2007. Marquard K , Westphal LM, Milki AA , Lathi RB. Etiology of recurrent pregnancy loss in women over the age of 35 years. Fertil Steril 94: 1473– 1477, 2010. Marques CJ, Costa P, Vaz B, et al. Abnormal methylation of imprinted genes in human sperm is associated with oligozoospermia. Mol Hum Reprod 14: 67–74, 2008. Marteau T, Drake H, Bobrow M. Counselling following diagnosis of a fetal abnormality: the differing approaches of obstetricians, clinical geneticists, and genetic nurses. J Med Genet 31: 864–867, 1994.
REFERENCES
•
571
Marteau TM, Dormandy E. Facilitating informed choice in prenatal testing: how well are we doing? Am J Med Genet 106: 185–190, 2001. Marteau TM, Nippert I, Hall S, et al. Outcomes of pregnancies diagnosed with Klinefelter syndrome: the possible influence of health professionals. Prenat Diagn 22: 562–566, 2002. Martens MA , Wilson SJ, Dudgeon P, Reutens DC. Approachability and the amygdala: insights from Williams syndrome. Neuropsychologia 47: 2446–2453, 2009. Martin CL, Nawaz Z, Baldwin EL, Wallace EJ, Justice AN, Ledbetter DH. The evolution of molecular ruler analysis for characterizing telomere imbalances: from fluorescence in situ hybridization to array comparative genomic hybridization. Genet Med 9: 566–573, 2007a. Martin JR , Wold A , Taylor HS. Ring chromosome 12 and severe oligospermia: a case report. Fertil Steril 90: 443 e13–15, 2008. Martin NDT, Smith WR , Cole TJ, Preece MA . New height, weight and head circumference charts for British children with Williams syndrome. Arch Dis Child 92: 598–601, 2007. Martin NJ, Cartwright DW, Harvey PJ. Duplication 5q(5q22→5q33): from an intrachromosomal insertion. Am J Med Genet 20: 57–62, 1985. Martin R . Human sperm chromosome complements in chemotherapy patients and infertile men. Chromosoma 107: 523–527, 1998. Martin RH. Sperm chromosome analysis in a man heterozygous for a paracentric inversion of chromosome 7 (q11q22). Hum Genet 73: 97–100, 1986. Martin RH. Sperm chromosome analysis in a man heterozygous for a paracentric inversion of chromosome 14 (q24.1q32.1). Am J Hum Genet 64: 1480–1484, 1999. Martin RH, Ernst S, Rademaker A , Barclay L, Ko E, Summers N. Chromosomal abnormalities in sperm from testicular cancer patients before and after chemotherapy. Hum Genet 99: 214–218, 1997. Martin RH, Ernst S, Rademaker A , Barclay L, Ko E, Summers N. Analysis of sperm chromosome complements before, during, and after chemotherapy. Cancer Genet Cytogenet 108: 133–136, 1999. Martin RH, Greene C, Rademaker A , Barclay L, Ko E, Chernos J. Chromosome analysis of spermatozoa extracted from testes of men with non-obstructive azoospermia. Hum Reprod 15: 1121–1124, 2000. Martin RH, Hildebrand K , Yamamoto J, et al. An increased frequency of human sperm
572
•
REFERENCES
chromosomal abnormalities after radiotherapy. Mutat Res 174: 219–225, 1986a. Martin RH, Hildebrand KA , Yamamoto J, et al. The meiotic segregation of human sperm chromosomes in two men with accessory marker chromosomes. Am J Med Genet 25: 381–388, 1986b. Martin RH, Ko E, Hildebrand K . Analysis of sperm chromosome complements from a man heterozygous for a robertsonian translocation 45,XY,t(15q;22q). Am J Med Genet 43: 855–857, 1992. Martin RH, Ko E, Rademaker A . Distribution of aneuploidy in human gametes: comparison between human sperm and oocytes. Am J Med Genet 39: 321–331, 1991. Martin RH, Rademaker A , German J. Chromosomal breakage in human spermatozoa, a heterozygous effect of the Bloom syndrome mutation. Am J Hum Genet 55: 1242–1246, 1994. Martinet D, Vial Y, Thonney F, Beckmann JS, Meagher-Villemure K , Unger S. Fetus with two identical reciprocal translocations: description of a rare complication of consanguinity. Am J Med Genet 140A : 769–774, 2006. Martínez JE, Tuck-Muller CM, Superneau D, Wertelecki W. Fertility and the cri du chat syndrome. Clin Genet 43: 212–214, 1993. Martinez-Castro P, Ramos MC, Rey JA , Benitez J, Sanchez Cascos A . Homozygosity for a Robertsonian translocation (13q14q) in three offspring of heterozygous parents. Cytogenet Cell Genet 38: 310–312, 1984. Martínez-Frías ML. The real earliest historical evidence of Down syndrome. Am J Med Genet 132A : 231, 2005. Martínez-Frías ML, Rodríguez-Pinilla E, Bermejo E, Prieto L. Epidemiological evidence that maternal diabetes does not appear to increase the risk for Down syndrome. Am J Med Genet 112: 335–337, 2002. Martínez-Pasarell O, Nogués C, Bosch M, Egozcue J, Templado C. Analysis of sex chromosome aneuploidy in sperm from fathers of Turner syndrome patients. Hum Genet 104: 345–349, 1999. Martinez-Pomar N, Munoz-Saa I, Heine-Suner D, Martin A , Smahi A , Matamoros N. A new mutation in exon 7 of NEMO gene: late skewed X-chromosome inactivation in an incontinentia pigmenti female patient with immunodeficiency. Hum Genet 118: 458–465, 2005. Mascarenhas A , Matoso E, Saraiva J, et al. First prenatally detected small supernumerary neocentromeric derivative chromosome 13
resulting in a non-mosaic partial tetrasomy 13q. Cytogenet Genome Res 121: 293–297, 2008. Mastenbroek S, Twisk M, van Echten-Arends J, et al. In vitro fertilization with preimplantation genetic screening. N Engl J Med 357: 9–17, 2007. Masuno M, Cholsong Y, Kuwahara T, et al. Second meiotic nondisjunction of the rearranged chromosome in a familial reciprocal 5/13 translocation. Am J Med Genet 41: 32–34, 1991. Masuno M, Imaizumi K , Ishii T, Kimura J, Kuroki Y. Supernumerary ring chromosome 5 identified by FISH. Am J Med Genet 84: 381, 1999. Masurel-Paulet A , Andrieux J, Callier P, et al. Delineation of 15q13.3 microdeletions. Clin Genet 78: 149–161, 2010. Mateizel I, Verheyen G, Van Assche E, Tournaye H, Liebaers I, Van Steirteghem A . FISH analysis of chromosome X, Y and 18 abnormalities in testicular sperm from azoospermic patients. Hum Reprod 17: 2249–2257, 2002. Matsui H, Iitsuka Y, Yamazawa K , Tanaka N, Seki K , Sekiya S. Changes in the incidence of molar pregnancies. A population-based study in Chiba Prefecture and Japan between 1974 and 2000. Hum Reprod 18: 172–175, 2003. Matsuo M, Muroya K , Kosaki K , et al. Random X-inactivation in a girl with duplication Xp11.21-p21. 3: report of a patient and review of the literature. Am J Med Genet 86: 44–50, 1999. Matsuo M, Muroya K , Nanao K , et al. Mother and daughter with 45,X/46,X,r(X)(p22.3q28) and mental retardation: analysis of the X-inactivation patterns. Am J Med Genet 91: 267–272, 2000. Mau Kai C, Juul A , McElreavey K , et al. Sons conceived by assisted reproduction techniques inherit deletions in the azoospermia factor (AZF) region of the Y chromosome and the DAZ gene copy number. Hum Reprod 23: 1669–1678, 2008. Maurin ML, Labrune P, Brisset S, et al. Molecular cytogenetic characterization of a 4p15.1-pter duplication and a 4q35.1-qter deletion in a recombinant of chromosome 4 pericentric inversion. Am J Med Genet 149A : 226–231, 2009. Mazzanti L, Cacciari E, Bergamaschi R , et al. Pelvic ultrasonography in patients with Turner syndrome: age-related findings in different karyotypes. J Pediatr 131: 135–140, 1997. McArthur SJ, Leigh D, Marshall JT, Gee AJ, De Boer KA , Jansen RPS. Blastocyst trophectoderm biopsy and preimplantation genetic diagnosis for familial monogenic disorders and chromosomal translocations. Prenat Diagn 28: 434–442, 2008. McClarren J, Donnenfeld AE, Ravnan JB. Prenatal diagnosis of an unexpected interstitial 22q11.2 deletion causing truncus arteriosus and thymic
hypoplasia in a ring 22 chromosome derived from a maternally inherited paracentric inversion. Prenat Diagn 26: 1212–1215, 2006. McConkie-Rosell A , Finucane B, Cronister A , Abrams L, Bennett RL, Pettersen BJ. Genetic counseling for fragile x syndrome: updated recommendations of the national society of genetic counselors. J Genet Couns 14: 249–270, 2005. McConkie-Rosell A , Spiridigliozzi GA , Rounds K , et al. Parental attitudes regarding carrier testing in children at risk for fragile X syndrome. Am J Med Genet 82: 206–211, 1999. McConkie-Rosell A , Spiridigliozzi GA , Sullivan JA , Dawson DV, Lachiewicz AM. Longitudinal study of the carrier testing process for fragile X syndrome: perceptions and coping. Am J Med Genet 98: 37–45, 2001. McConkie-Rosell A , Spiridigliozzi GA , Sullivan JA , Dawson DV, Lachiewicz AM. Carrier testing in fragile X syndrome: when to tell and test. Am J Med Genet 110: 36–44, 2002. McDermid HE, Morrow BE. Genomic disorders on 22q11. Am J Hum Genet 70: 1077–1088, 2002. McDermott EM, Powell RJ. Incidence of ovarian failure in systemic lupus erythematosus after treatment with pulse cyclophosphamide. Ann Rheum Dis 55: 224–229, 1996. McFadden DE, Friedman JM. Chromosome abnormalities in human beings. Mutat Res 396: 129–140, 1997. McFadden DE, Jiang R , Langlois S, Robinson WP. Dispermy—origin of diandric triploidy: brief communication. Hum Reprod 17: 3037–3038, 2002. McFadden DE, Kwong LC, Yam IY, Langlois S. Parental origin of triploidy in human fetuses: evidence for genomic imprinting. Hum Genet 92: 465–469, 1993. McFadden DE, Langlois S. Parental and meiotic origin of triploidy in the embryonic and fetal periods. Clin Genet 58: 192–200, 2000. McFadden DE, Robinson WP. Phenotype of triploid embryos. J Med Genet 43: 609–612, 2006. McGaughran J, Hadwen T, Clark R . Progressive edema leading to pleural effusions in a female with a ring chromosome 22 leading to a 22q13 deletion. Clin Dysmorphol 19: 28–29, 2010. McGhee EM, Klump CJ, Bitts SM, Cotter PD, Lammer EJ. Candidate region for Coffin-Siris syndrome at 7q32→34. Am J Med Genet 93: 241–243, 2000. McGoey RR , Lacassie Y. Paternal balanced reciprocal translocation t(9;22)(q34.3;q11.2) resulting in an infant with features of the 9q subtelomere and the 22q11 deletion syndromes due to 3:1 meiotic
REFERENCES
•
573
segregation and tertiary monosomy. Am J Med Genet 149A : 2538–2542, 2009. McGowan KD, Weiser JJ, Horwitz J, et al. The importance of investigating for uniparental disomy in prenatally identified balanced acrocentric rearrangements. Prenat Diagn 22: 141–143, 2002. McKenzie LJ, Cisneros PL, Torsky S, et al. Preimplantation genetic diagnosis for a known cryptic translocation: follow-up clinical report and implication of segregation products. Am J Med Genet 121A : 56–59, 2003. McWeeney DT, Munné S, Miller RC, et al. Pregnancy complicated by triploidy: a comparison of the three karyotypes. Am J Perinatol 26: 641–645, 2009. Mears AJ, Duncan AMV, Budarf ML, et al. Molecular characterization of the marker chromosome associated with cat eye syndrome. Am J Hum Genet 55: 134–142, 1994. Mears AJ, el-Shanti H, Murray JC, McDermid HE, Patil SR . Minute supernumerary ring chromosome 22 associated with cat eye syndrome: further delineation of the critical region. Am J Hum Genet 57: 667–673, 1995. Mefford HC, Sharp AJ, Baker C, et al. Recurrent rearrangements of chromosome 1q21.1 and variable pediatric phenotypes. N Engl J Med 359: 1685–1699, 2008. Mehra S, Christ L, Jeng L, Zinn AB, Schwartz S. Characterization of a familial balanced rec(13) in a child with mild MR and his half-sibling with two structurally rearranged chromosomes 13. Am J Med Genet 137: 217–221, 2005. Mehraein Y, Wippermann CF, Michel-Behnke I, et al. Microdeletion 22q11 in complex cardiovascular malformations. Hum Genet 99: 433–442, 1997. Meiner A , Holland H, Reichenbach H, Horn LC, Faber R , Froster UG. Tetraploidy in a growthretarded fetus with a thick placenta. Prenat Diagn 18: 864–865, 1998. Meistrich ML, Byrne J. Genetic disease in offspring of long-term survivors of childhood and adolescent cancer treated with potentially mutagenic therapies. Am J Hum Genet 70: 1069–1071, 2002. Melichar VO, Guth S, Hellebrand H, et al. A male infant with a 9.6 Mb terminal Xp deletion including the OA1 locus: Limit of viability of Xp deletions in males. Am J Med Genet 143: 135–141, 2007. Melnyk AR , Ahmed I, Taylor JC. Prenatal diagnosis of familial ring 21 chromosome. Prenat Diagn 15: 269–273, 1995. Melotte C, Debrock S, D’Hooghe T, Fryns JP, Vermeesch JR . Preimplantation genetic diagnosis
574
•
REFERENCES
for an insertional translocation carrier. Hum Reprod 19: 2777–2783, 2004. Menasha J, Levy B, Hirschhorn K , Kardon NB. Incidence and spectrum of chromosome abnormalities in spontaneous abortions: new insights from a 12-year study. Genet Med 7: 251–263, 2005. Mencarelli MA , Katzaki E, Papa FT, et al. Private inherited microdeletion/microduplications: implications in clinical practice. Eur J Med Genet 51: 409–416, 2008. Mendes JRT, Strufaldi MWL, Delcelo R , et al. Y-chromosome identification by PCR and gonadal histopathology in Turner’s syndrome without overt Y-mosaicism. Clin Endocrinol 50: 19–26, 1999. Menon S, Rives N, Mousset-Siméon N, et al. Fertility preservation in adolescent males: experience over 22 years at Rouen University Hospital. Hum Reprod 24: 37–44, 2009. Menten B, Maas N, Thienpont B, et al. Emerging patterns of cryptic chromosomal imbalance in patients with idiopathic mental retardation and multiple congenital anomalies: a new series of 140 patients and review of published reports. J Med Genet 43: 625–633, 2006. Mercier S, Bresson JL. Analysis of chromosomal equipment in spermatozoa of a 46,XY/47,XY/+8 male by means of multicolour fluorescent in situ hybridization: confirmation of a mosaicism and evaluation of risk for offspring. Hum Genet 99: 42–46, 1997. Merrill A , Rosenblum-Vos L, Driscoll DA , Daley K , Treat K . Prenatal diagnosis of Fanconi anemia (Group C) subsequent to abnormal sonographic findings. Prenat Diagn 25: 20–22, 2005. Meschede D, Froster UG, Bergmann M, Nieschlag E. Familial pericentric inversion of chromosome 1 (p34q23) and male infertility with stage specific spermatogenic arrest. J Med Genet 31: 573–575, 1994. Meschede D, Lemcke B, Exeler JR , et al. Chromosome abnormalities in 447 couples undergoing intracytoplasmic sperm injection— prevalence, types, sex distribution and reproductive relevance. Hum Reprod 13: 576–582, 1998a. Meschede D, Lemcke B, Stussel J, Louwen F, Horst J. Strong preference for non-invasive prenatal diagnosis in women pregnant through intracytoplasmic sperm injection (ICSI). Prenat Diagn 18: 700–705, 1998b. Meschede D, Louwen F, Eiben B, Horst J. Intracytoplasmic sperm injection pregnancy with fetal trisomy 9p resulting from a balanced paternal translocation. Hum Reprod 12: 1913–1914, 1997.
Meschede D, Louwen F, Nippert I, Holzgreve W, Miny P, Horst J. Low rates of pregnancy termination for prenatally diagnosed Klinefelter syndrome and other sex chromosome polysomies. Am J Med Genet 80: 330–334, 1998c. Metcalfe K , Simeonov E, Beckett W, Donnai D, Tassabehji M. Autosomal dominant inheritance of Williams-Beuren syndrome in a father and son with haploinsufficiency for FKBP6. Clin Dysmorphol 14: 61–65, 2005. Meyer E, Lim D, Pasha S, et al. Germline mutation in NLRP2 (NALP2) in a familial imprinting disorder (Beckwith-Wiedemann Syndrome). PLoS Genet 5: e1000423, 2009. Micale M, Insko J, Ebrahim SAD, Adeyinka A , Runke C, Van Dyke DL. Double trisomy revisited—a multicenter experience. Prenat Diagn 30: 173–176, 2010. Michaelis RC, Velagaleti GVN, Jones C, et al. Most Jacobsen syndrome deletion breakpoints occur distal to FRA11B. Am J Med Genet 76: 222–228, 1998. Michelson RJ, Weinert T. Closing the gaps among a web of DNA repair disorders. Bioessays 22: 966–969, 2000. Michie S, Bobrow M, Marteau TM. Predictive genetic testing in children and adults: a study of emotional impact. J Med Genet 38: 519–526, 2001. Middleton FA , Trauzzi MG, Shrimpton AE, et al. Complete maternal uniparental isodisomy of chromosome 4 in a subject with major depressive disorder detected by high density SNP genotyping arrays. Am J Med Genet 141B: 28–32, 2006. Midro AT, Panasiuk B, Stasiewicz-Jarocka B, et al. Risk estimates for carriers of chromosome reciprocal translocation t(4;9)(p15.2;p13). Clin Genet 58: 153–155, 2000. Midro AT, Stengel-Rutkowski S, Stene J. Experiences with risk estimates for carriers of chromosomal reciprocal translocations. Clin Genet 41: 113–122, 1992. Midro AT, Wiland E, Panasiuk B, Lesniewicz R , Kurpisz M. Risk evaluation of carriers with chromosome reciprocal translocation t(7;13) (q34;q13) and concomitant meiotic segregation analyzed by FISH on ejaculated spermatozoa. Am J Med Genet 140: 245–256, 2006. Migeon B. Females Are Mosaics: X Inactivation and Sex Differences in Disease. New York: Oxford University Press, 2007. Migeon BR , Ausems M, Giltay J, et al. Severe phenotypes associated with inactive ring X chromosomes. Am J Med Genet 93: 52–57, 2000. Mikhaail-Philips MM, Ko E, Chernos J, Greene C, Rademaker A , Martin RH. Analysis of
chromosome segregation in sperm from a chromosome 2 inversion heterozygote and assessment of an interchromosomal effect. Am J Med Genet 127A : 139–143, 2004. Mikhaail-Philips MM, McGillivray BC, Hamilton SJ, et al. Unusual segregation products in sperm from a pericentric inversion 17 heterozygote. Hum Genet 117: 357–365, 2005. Mikkelsen M. Familial Down’s syndrome. A cytogenetical and genealogical study of twentytwo families. Ann Hum Genet 30: 125–146, 1966. Milazzo JP, Rives N, Mousset-Siméon N, Macé B. Chromosome constitution and apoptosis of immature germ cells present in sperm of two 47,XYY infertile males. Hum Reprod 21: 1749– 1758, 2006. Miller DT, Adam MP, Aradhya S, et al. Consensus statement: chromosomal microarray is a first-tier clinical diagnostic test for individuals with developmental disabilities or congenital anomalies. Am J Hum Genet 86: 749–764, 2010. Miller DT, Shen Y, Weiss LA , et al. Microdeletion/ duplication at 15q13.2q13.3 among individuals with features of autism and other neuropsychiatric disorders. J Med Genet 46: 242–248, 2009. Miller JF, Williamson E, Glue J, Gordon YB, Grudzinskas JG, Sykes A . Fetal loss after implantation. A prospective study. Lancet 2: 554–556, 1980. Miller K , Müller W, Winkler L, Hadam MR , Ehrich JHH, Flatz SD. Mitotic disturbance associated with mosaic aneuploidies. Hum Genet 84: 361–364, 1990. Miller K , Pabst B, Ritter H, et al. Chromosome 18 replaced by two ring chromosomes of chromosome 18 origin. Hum Genet 112: 343–347, 2003. Miller K , Reimer A , Schulze B. Tandem duplication chromosome 21 in the offspring of a ring chromosome 21 carrier. Ann Génét 30: 180–182, 1987. Miller KR , Mühlhaus K , Herbst RA , Bohnhorst B, Böhmer S, Arslan-Kirchner M. Patient with trisomy 6 mosaicism. Am J Med Genet 100: 103–105, 2001. Miller OJ, Therman E. Human Chromosomes. 4th ed. New York: Springer, 2001. Miller VL, Ransom SB, Ayoub MA , Krivchenia EL, Evans MI. Fiscal impact of a potential legislative ban on second trimester elective terminations for prenatally diagnosed abnormalities. Am J Med Genet 91: 359–362, 2000. Ming JE, Blagowidow N, Knoll JHM, et al. Submicroscopic deletion in cousins with Prader-Willi syndrome causes a grandmatrilineal
REFERENCES
•
575
inheritance pattern: effects of imprinting. Am J Med Genet 92: 19–24, 2000. Minor A , Harmer K , Peters N, Yuen BH, Ma S. Investigation of confined placental mosaicism (CPM) at multiple sites in post-delivery placentas derived through intracytoplasmic sperm injection (ICSI). Am J Med Genet 140: 24–30, 2006. Minto CL, Crouch NS, Conway GS, Creighton SM. XY females: revisiting the diagnosis. Br J Obstet Gynaecol 112: 1407–1410, 2005. Missirian C, Moncla A , Voelckel MA , Ravix V, Philip N. Fragile X syndrome and 22q11.2 microdeletion in the same sibship. Am J Med Genet 95: 358–360, 2000. Mitchell JJ, Vekemans M, Luscombe S, et al. U-type exchange in a paracentric inversion as a possible mechanism of origin of an inverted tandem duplication of chromosome 8. Am J Med Genet 49: 384–387, 1994. Mitelman F, Johansson B, Mertens F (Eds.). Mitelman Database of Chromosome Aberrations and Gene Fusions in Cancer. http://cgap.nci.nih.gov/ Chromosomes/Mitelman, 2011. Mitra A , Dada R , Kumar R , Gupta NP, Kucheria K , Gupta SK . Y chromosome microdeletions in azoospermic patients with Klinefelter’s syndrome. Asian J Androl 8: 81–88, 2006. Mitter D, Buiting K , von Eggeling F, et al. Is there a higher incidence of maternal uniparental disomy 14 [upd(14)mat]? Detection of 10 new patients by methylation-specific PCR . Am J Med Genet 140: 2039–2049, 2006. Mittwoch U. Sex determination and sex reversal: genotype, phenotype, dogma and semantics. Hum Genet 89: 467–479, 1992. Miura K , Obama M, Yun K , et al. Methylation imprinting of H19 and SNRPN genes in human benign ovarian teratomas. Am J Hum Genet 65: 1359–1367, 1999. Miura Y, Hiura M, Torigoe K , et al. Complete paternal uniparental isodisomy for chromosome 1 revealed by mutation analyses of the TRKA (NTRK1) gene encoding a receptor tyrosine kinase for nerve growth factor in a patient with congenital insensitivity to pain with anhidrosis. Hum Genet 107: 205–209, 2000. Miyamoto T, Hasuike S, Yogev L, et al. Azoospermia in patients heterozygous for a mutation in SYCP3. Lancet 362: 1714–1719, 2003. Miyoshi O, Yabe R , Wakui K , et al. Two cases of mosaic RhD blood-group phenotypes and paternal isodisomy for chromosome 1. Am J Med Genet 104: 250–256, 2001. Mizutani E, Suzumori N, Ozaki Y, et al. SYCP3 mutation may not be associated with recurrent
576
•
REFERENCES
miscarriage caused by aneuploidy. Hum Reprod 26: 1259–66, 2011. Moayeri SE, Allen RB, Brewster WR , Kim MH, Porto M, Werlin LB. Day-3 embryo morphology predicts euploidy among older subjects. Fertil Steril 89: 118–123, 2008. Mochizuki J, Saitsu H, Mizuguchi T, et al. Alu-related 5q35 microdeletions in Sotos syndrome. Clin Genet 74: 384–391, 2008. Modan-Moses D, Litmanovitch T, Rienstein S, Meyerovitch J, Goldman B, Aviram-Goldring A . True hermaphroditism with ambiguous genitalia due to a complicated mosaic karyotype: clinical features, cytogenetic findings, and literature review. Am J Med Genet 116A : 300–303, 2003. Modi DN, Sane S, Bhartiya D. Accelerated germ cell apoptosis in sex chromosome aneuploid fetal human gonads. Mol Hum Reprod 9: 219–225, 2003. Mokate T, Leask K , Mehta S, et al. Non-mosaic trisomy 22: a report of 2 cases. Prenat Diagn 26: 962–965, 2006. Molina O, Blanco J, Vidal F. Deletions and duplications of the 15q11-q13 region in spermatozoa from Prader-Willi syndrome fathers. Mol Hum Reprod 16: 320–328, 2010. Molina O, Anton E, Vidal F, Blanco J. Sperm rates of 7q11.23, 15q11q13 and 22q11.2 deletions and duplications: a FISH approach. Hum Genet 129: 35–44, 2011. Molina Gomes D, Hammoud I, Bailly M, et al. Preconceptional diagnosis for Robertsonian translocation as an alternative to preimplantation genetic diagnosis in two situations: a pilot study. J Assist Reprod Genet 26: 113–117, 2009. Momeni P, Glöckner G, Schmidt O, et al. Mutations in a new gene, encoding a zinc-finger protein, cause tricho-rhino-phalangeal syndrome type I. Nat Genet 24: 71–74, 2000. Monahan P. Judgements of interest: A doctor’s dilemma resolved—sterilisation of intellectually disabled females. Australasian J Med Defence Union 3: 54–55, 1992. Moncla A , Malzac P, Livet MO, et al. Angelman syndrome resulting from UBE3A mutations in 14 patients from eight families: clinical manifestations and genetic counselling. J Med Genet 36: 554–560, 1999. Monni G, Cau G, Lai R , Demontis G, Usai V. Intracytoplasmic sperm injection and prenatal invasive diagnosis. Prenat Diagn 19: 390, 1999. Monnot S, Giuliano F, Massol C, et al. Partial Xp11.23-p11.4 duplication with random X inactivation: clinical report and molecular
cytogenetic characterization. Am J Med Genet 146A : 1325–1329, 2008. Montag M, van der Ven K , Rösing B, van der Ven H. Polar body biopsy: a viable alternative to preimplantation genetic diagnosis and screening. Reprod Biomed Online 18(suppl 1): 6–11, 2009. Moore CJ, Daly EM, Schmitz N, et al. A neuropsychological investigation of male premutation carriers of fragile X syndrome. Neuropsychologia 42: 1934–1947, 2004. Moore GE, Ali Z, Khan RU, Blunt S, Bennett PR , Vaughan JI. The incidence of uniparental disomy associated with intrauterine growth retardation in a cohort of thirty-five severely affected babies. Am J Obstet Gynecol 176: 294–299, 1997. Moradkhani K , Puechberty J, Bhatt S, et al. Rare Robertsonian translocations and meiotic behaviour: sperm FISH analysis of t(13;15) and t(14;15) translocations: a case report. Hum Reprod 21: 3193–3198, 2006a. Moradkhani K , Puechberty J, Bhatt S, et al. Meiotic segregation of rare Robertsonian translocations: sperm analysis of three t(14q;22q) cases. Hum Reprod 21: 1166–1171, 2006b. Morales C, Madrigal I, Esqué T, et al. Duplication/ deletion mosaicism of the 7q(21.1 → 31.3) region. Am J Med Genet 143: 179–183, 2007a. Morales C, Sánchez A , Bruguera J, et al. Cytogenetic study of spontaneous abortions using semi-direct analysis of chorionic villi samples detects the broadest spectrum of chromosome abnormalities. Am J Med Genet 146A : 66–70, 2008. Morales C, Soler A , Bruguera J, et al. Pseudodicentric 22;Y translocation transmitted through four generations of a large family without phenotypic repercussion. Cytogenet Genome Res 116: 319–323, 2007b. Morel F, Duguépéroux I, McElreavey K , et al. Transmission of an unbalanced (Y;1) translocation in Brittany, France. J Med Genet 39: e52, 2002a. Morel F, Fellmann F, Roux C, Bresson JL. Meiotic segregation analysis by FISH investigation of spermatozoa of a 46,Y,der(X),t(X;Y)(qter→p 22::q11→qter) carrier. Cytogenet Cell Genet 92: 63–68, 2001. Morel F, Gallon F, Amice V, et al. Sex chromosome mosaicism in couples undergoing intracytoplasmic sperm injection. Hum Reprod 17: 2552–2555, 2002b. Morel F, Laudier B, Guérif F, et al. Meiotic segregation analysis in spermatozoa of pericentric inversion carriers using fluorescence in-situ hybridization. Hum Reprod 22: 136–141, 2007. Morel Y, Mebarki F, Forest MG. What are the indications for prenatal diagnosis in the androgen
insensitivity syndrome? Facing clinical heterogeneity of phenotypes for the same genotype. Eur J Endocrinol 130: 325–326, 1994. Mori MA , Huertas H, Pinel I, Giralt P, Martínez-Frías ML. Trisomy 13 in the child of two carriers of a 13/15 translocation. Am J Med Genet 20: 17–20, 1985. Morichon-Delvallez N, Segues B, Pinson MP, et al. Maternal uniparental disomy for chromosome 14 by secondary nondisjunction of an initial trisomy. Am J Hum Genet 55(suppl): 379A , 1994. Morris A , Boyd E, Dhanjal S, et al. Two years’ prospective experience using fluorescence in situ hybridization on uncultured amniotic fluid cells for rapid prenatal diagnosis of common chromosomal aneuploidies. Prenat Diagn 19: 546–551, 1999. Morris JK , Alberman E. Trends in Down’s syndrome live births and antenatal diagnoses in England and Wales from 1989 to 2008: analysis of data from the National Down Syndrome Cytogenetic Register. Brit Med J 339: b3794, 2009 Morris JK , Alberman E, Scott C, Jacobs P. Is the prevalence of Klinefelter syndrome increasing? Eur J Hum Genet 16: 163–170, 2008. Morris JK , De Vigan C, Mutton DE, Alberman E. Risk of a Down syndrome live birth in women 45 years of age and older. Prenat Diagn 25: 275–278, 2005a. Morris JK , Mutton DE, Alberman E. Revised estimates of the maternal age specific live birth prevalence of Down’s syndrome. J Med Screen 9: 2–6, 2002. Morris JK , Mutton DE, Alberman E. Recurrences of free trisomy 21: analysis of data from the National Down Syndrome Cytogenetic Register. Prenat Diagn 25: 1120–1128, 2005b. Morris JK , Wald NJ, Mutton DE, Alberman E. Comparison of models of maternal age-specific risk for Down syndrome live births. Prenat Diagn 23: 252–258, 2003. Mortimer JG, Chewings WE, Gardner RJM. A further report on a kindred with cases of 4p trisomy and monosomy. Hum Hered 30: 58–61, 1980. Mroz K , Hassold TJ, Hunt PA . Meiotic aneuploidy in the XXY mouse: evidence that a compromised testicular environment increases the incidence of meiotic errors. Hum Reprod 14: 1151–1156, 1999. Muhlhauser A , Susiarjo M, Rubio C, et al. Bisphenol A effects on the growing mouse oocyte are influenced by diet. Biol Reprod 80: 1066–1071, 2009. Mulcahy MT, Jenkyn J, Mackellar A . 45,X/47,XYY mosaicism. J Med Genet 14: 218–221, 1977. Mules EH, Stamberg J. Reproductive outcomes of paracentric inversion carriers: report of a liveborn
REFERENCES
•
577
dicentric recombinant and literature review. Hum Genet 67: 126–131, 1984. Muller F, Dreux S, Dupoizat H, et al. Second-trimester Down syndrome maternal serum screening in twin pregnancies: impact of chorionicity. Prenat Diagn 23: 331–335, 2003. Muller F, Rebiffé M, Taillandier A , Oury JF, Mornet E. Parental origin of the extra chromosome in prenatally diagnosed fetal trisomy 21. Hum Genet 106: 340–344, 2000. Müller J, Ritzén EM, Ivarsson SA , Rajpert-De Meyts E, Norjavaara E, Skakkebæk NE. Management of males with 45,X/46,XY gonadal dysgenesis. Horm Res 52: 11–14, 1999. Müller J, Skakkebæk NE. Gonadal malignancy in individuals with sex chromosome anomalies. Birth Defects Orig Artic Ser 26: 247–255, 1990. Mulley JC, Yu S, Loesch DZ, et al. FRAXE and mental retardation. J Med Genet 32: 162–169, 1995. Munné S. Analysis of chromosome segregation during preimplantation genetic diagnosis in both male and female translocation heterozygotes. Cytogenet Genome Res 111: 305–309, 2005. Munné S, Bahçe M, Schimmel T, Sadowy S, Cohen J. Case report: chromatid exchange and predivision of chromatids as other sources of abnormal oocytes detected by preimplantation genetic diagnosis of translocations. Prenat Diagn 18: 1450–1458, 1998a. Munné S, Cohen J, Sable D. Preimplantation genetic diagnosis for advanced maternal age and other indications. Fertil Steril 78: 234–236, 2002a. Munné S, Escudero T, Fischer J, et al. Negligible interchromosomal effect in embryos of Robertsonian translocation carriers. Reprod Biomed Online 10: 363–369, 2005. Munné S, Escudero T, Sandalinas M, Sable D, Cohen J. Gamete segregation in female carriers of Robertsonian translocations. Cytogenet Cell Genet 90: 303–308, 2000a. Munné S, Morrison L, Fung J, et al. Spontaneous abortions are reduced after preconception diagnosis of translocations. J Assit Reprod Genet 15: 290–296, 1998b. Munné S, Sandalinas M, Escudero T, Fung J, Gianaroli L, Cohen J. Outcome of preimplantation genetic diagnosis of translocations. Fertil Steril 73: 1209–1218, 2000b. Munné S, Sandalinas M, Escudero T, Marquez C, Cohen J. Chromosome mosaicism in cleavagestage human embryos: evidence of a maternal age effect. Reprod Biomed Online 4: 223–232, 2002b. Munné S, Sandalinas M, Magli C, Gianaroli L, Cohen J, Warburton D. Increased rate of aneuploid
578
•
REFERENCES
embryos in young women with previous aneuploid conceptions. Prenat Diagn 24: 638–643, 2004. Muroya K , Yamamoto K , Fukushima Y, Ogata T. Ring chromosome 21 in a boy and a derivative chromosome 21 in the mother: implication for ring chromosome formation. Am J Med Genet 110: 332–337, 2002. Murphy EA , Chase GA . Principles of Genetic Counseling. Chicago: Year Book Medical Publishers, 1975. Murray J, Cuckle H, Sehmi I, Wilson C, Ellis A . Quality of written information used in Down syndrome screening. Prenat Diagn 21: 138–142, 2001. Musio A , Selicorni A , Focarelli ML, et al. X-linked Cornelia de Lange syndrome owing to SMC1L1 mutations. Nat Genet 38: 528–530, 2006. Mutter M, Binkert F, Schinzel A . Down syndrome livebirth rate in the eastern part of Switzerland between 1980 and 1996 stays constant in spite of growing numbers of prenatally diagnosed and subsequently terminated cases. Prenat Diagn 22: 835–836, 2002. Nadal M, Moreno S, Pritchard M, Preciado MA , Estivill X , Ramos-Arroyo MA . Down syndrome: characterisation of a case with partial trisomy of chromosome 21 owing to a paternal balanced translocation (15;21) (q26; q22.1) by FISH. J Med Genet 34: 50–54, 1997. Nadal M, Valiente A , Domènech A , Pritchard M, Estivill X , Ramos-Arroyo MA . Hereditary neuropathy with liability to pressure palsies: two cases with a reciprocal translocation t(16;17) (q12;11.2) interrupting the PMP22 gene. J Med Genet 37: 396–398, 2000. Nagai T, Shimokawa O, Harada N, et al. Postnatal overgrowth by 15q-trisomy and intrauterine growth retardation by 15q-monosomy due to familial translocation t(13;15): dosage effect of IGF1R? Am J Med Genet 113: 173–177, 2002. Nagamani SCS, Zhang F, Shchelochkov OA , et al. Microdeletions including YWHAE in the Miller-Dieker syndrome region on chromosome 17p13.3 result in facial dysmorphisms, growth restriction, and cognitive impairment. J Med Genet 46: 825–833, 2009. Nagarsheth NP, Mootabar H. Prenatal diagnosis of mosaic tetrasomy 21q confirmed by fluorescence in situ hybridization. Clin Genet 51: 260–263, 1997. Nagle C, Hodges R , Wolfe R , Wallace EM. Reporting Down syndrome screening results: women’s understanding of risk . Prenat Diagn 29: 234–239, 2009.
Nakamura N. Genetic effects of radiation in atomicbomb survivors and their children: past, present and future. J Radiat Res 47: B67–73, 2006. Nanal R , Kyle P, Soothill PW. A classification of pregnancy losses after invasive prenatal diagnostic procedures: an approach to allow comparison of units with a different case mix. Prenat Diagn 23: 488–492, 2003. Nap AW, Van Golde RJ, Tuerlings JH, et al. Reproductive decisions of men with microdeletions of the Y chromosome: the role of genetic counselling. Hum Reprod 14: 2166–2169, 1999. Narchi H, Kulaylat N. High incidence of Down’s syndrome in infants of diabetic mothers. Arch Dis Child 77: 242–244, 1997. Nardmann J, Tranebjærg L, Horsthemke B, Lüdecke HJ. The tricho-rhino-phalangeal syndromes: frequency and parental origin of 8q deletions. Hum Genet 99: 638–643, 1997. Natacci F, Corrado L, Pierri M, et al. Patient with large 17p11.2 deletion presenting with Smith-Magenis syndrome and Joubert syndrome phenotype. Am J Med Genet 95: 467–472, 2000. Navarro J, Vidal F, Benet J, Templado C, Marina S, Egozcue J. XY-trivalent association and synaptic anomalies in a male carrier of a Robertsonian t(13;14) translocation. Hum Reprod 6: 376–381, 1991. Neal MS, Cowan L, Louis JP, Hughes E, King WA , Basrur PK . Cytogenetic evaluation of human oocytes that failed to complete meiotic maturation in vitro. Fertil Steril 77: 844–845, 2002. Neas KR , Yip MY, James C, Kirk EPE. Patient with a non-mosaic isodicentric Yp and mild developmental delay. Am J Med Genet 137: 223–224, 2005. Neel JV, Schull WJ. The Children of Atomic Bomb Survivors: A Genetic Study. Washington, DC: National Academy Press, 1991. Neel JV, Schull WJ, Awa AA , et al. The children of parents exposed to atomic bombs: estimates of the genetic doubling dose of radiation for humans. Am J Hum Genet 46: 1053–1072, 1990. Neiswanger K , Hohler PM, Hively-Thomas LB, McPherson EW, Hogge WA , Surti U. Variable outcomes in mosaic trisomy 16: five case reports and literature analysis. Prenat Diagn 26: 454–461, 2006. Neitzel H, Neumann LM, Schindler D, et al. Premature chromosome condensation in humans associated with microcephaly and mental retardation: a novel autosomal recessive condition. Am J Hum Genet 70: 1015–1022, 2002.
Nelis E, Timmerman V, De Jonghe P, Van Broeckhoven C, Rautenstrauss B. Molecular genetics and biology of inherited peripheral neuropathies: a fast-moving field. Neurogenet 2: 137–148, 1999. Németh AH, Gallen IW, Crocker M, Levy E, Maher E. Klinefelter-like phenotype and primary infertility in a male with a paracentric Xq inversion. J Med Genet 39: E28, 2002. Neri G. A possible explanation for the low incidence of gonosomal aneuploidy among the offspring of triplo-X individuals. Am J Med Genet 18: 357–364, 1984. Neri G, Opitz JM. Down syndrome: comments and reflections on the 50th anniversary of Lejeune’s discovery. Am J Med Genet 149A : 2647–2654, 2009. Neri G, Romana Di Raimo F. Long time no see: the type and contre-type concept. Eur J Hum Genet 18: 135–136, 2010. Nerlich A , Wisser J, Krone S. Placental findings in “vanishing twins.” Geburtshilfe Frauenheilkd 52: 230–234, 1992. Ness GO, Lybæk H, Houge G. Usefulness of high-resolution comparative genomic hybridization (CGH) for detecting and characterizing constitutional chromosome abnormalities. Am J Med Genet 113: 125–136, 2002. Netley CT. Summary overview of behavioural development in individuals with neonatally identified X and Y aneuploidy. Birth Defects Orig Artic Ser 22: 293–306, 1986. Netzer C, Klein C, Kohlhase J, Kubisch C. New challenges for informed consent through whole genome array testing. J Med Genet 46: 495–496, 2009. Neu RL, Brar HS, Koos BJ. Prenatal diagnosis of inv(X)(q12q28) in a male fetus. J Med Genet 25: 52–53, 1988a. Neu RL, Kousseff BG, Hardy DE, et al. Trisomy 3p23→pter and monosomy 11q23→qter in an infant with two translocation carrier parents. J Med Genet 25: 631–633, 1988b. Neu RL, Valentine FA , Gardner LI. Segregation of a t(14q22q) chromosome in a large kindred. Clin Genet 8: 30–36, 1975. Neumann AA , Robson LG, Smith A . A 15p+ variant shown to be a t(Y;15) with fluorescence in situ hybridisation. Ann Génét 35: 227–230, 1992. Nevado J, de Torres ML, Fernández L, et al. Unusual four-generation chromosome-22 rearrangement: when “normality” masks abnormality. Am J Med Genet 149A : 1561–1564, 2009.
REFERENCES
•
579
Newberg MT, Francisco RG, Pang MG, et al. Cytogenetics of somatic cells and sperm from a 46,XY/45,X mosaic male with moderate oligoasthenoteratozoospermia. Fertil Steril 69: 146–148, 1998. Ngo AD, Taylor R , Roberts CL, Nguyen TV. Association between Agent Orange and birth defects: systematic review and meta-analysis. Int J Epidemiol 35: 1220–1230, 2006. Nicholson HS, Byrne J. Fertility and pregnancy after treatment for cancer during childhood or adolescence. Cancer 71: 3392–3399, 1993. Nickerson E, Greenberg F, Keating MT, McCaskill C, Shaffer LG. Deletions of the elastin gene at 7q11.23 occur in approximately 90% of patients with Williams syndrome. Am J Hum Genet 56: 1156–1161, 1995. Nicolaides K , Brizot Mde L, Patel F, Snijders R . Comparison of chorionic villus sampling and amniocentesis for fetal karyotyping at 10–13 weeks’ gestation. Lancet 344: 435–439, 1994. Nicolaidis P, Petersen MB. Origin and mechanisms of non-disjunction in human autosomal trisomies. Hum Reprod 13: 313–319, 1998. Nicopoullos JDM, Gilling-Smith C, Almeida PA , et al. The role of sperm aneuploidy as a predictor of the success of intracytoplasmic sperm injection? Hum Reprod 23: 240–250, 2008. Nicosia SV, Matus-Ridley M, Meadows AT. Gonadal effects of cancer therapy in girls. Cancer 55: 2364–2372, 1985. Niedrist D, Lurie IW, Schinzel A . 4q32-q35 and 6q16-q22 are valuable candidate regions for split hand/foot malformation. Eur J Hum Genet 17: 1086–1091, 2009. Nielsen J. Follow-up of 25 unselected children with sex chromosome abnormalities to age 12. Birth Defects Orig Artic Ser 26: 201–207, 1990. Nielsen J, Wohlert M. Chromosome abnormalities found among 34,910 newborn children: results from a 13-year incidence study in Arhus, Denmark . Hum Genet 87: 81–83, 1991. Nielsen J, Wohlert M, Faaborg-Andersen J, et al. (1986). Chromosome examination of 20,222 newborn children: results from a 7.5-year study in Århus, Denmark . Birth Defects Orig Art Series 22(3): 209–219, 1986. Nielsen KG, Poulsen H, Mikkelsen M, Steuber E. Multiple recurrence of trisomy 21 Down syndrome. Hum Genet 78: 103–105, 1988. Niessen RC, Jonkman MF, Muis N, Hordijk R , van Essen AJ. Pigmentary mosaicism following the lines of Blaschko in a girl with a double aneuploidy mosaicism: (47,XX,+7/45,X). Am J Med Genet 137A : 313–322, 2005.
580
•
REFERENCES
Nieuwint A , Van Zalen-Sprock R , Hummel P, et al. “Identical” twins with discordant karyotypes. Prenat Diagn 19: 72–76, 1999. Nikolaou D, Templeton A . Early ovarian ageing. Eur J Obstet Gynecol Reprod Biol 113: 126–133, 2004. Niu DM, Huang JY, Li HY, et al. Paternal gonadal mosaicism of NIPBL mutation in a father of siblings with Cornelia de Lange syndrome. Prenat Diagn 26: 1054–1057, 2006. Niu DM, Pan CC, Lin CY, Hwang B, Chung MY. Mosaic or chimera? Revisiting an old hypothesis about the cause of the 46,XX/46,XY hermaphrodite. J Pediatr 140: 732–735, 2002. Nolin SL, Brown WT, Glicksman A , et al. Expansion of the fragile X CGG repeat in females with premutation or intermediate alleles. Am J Hum Genet 72: 454–464, 2003. Nolin SL, Glicksman A , Ding X, et al. Fragile X analysis of 1112 prenatal samples from 1991 to 2010. Prenat Diagn, in press (published online June 2011). Norman AM, Read AP, Clayton-Smith J, Andrews T, Donnai D. Recurrent Wiedemann-Beckwith syndrome with inversion of chromosome (11) (p11.2p15.5). Am J Med Genet 42: 638–641, 1992. Northrop LE, Treff NR , Levy B, Scott RT. SNP microarray-based 24 chromosome aneuploidy screening demonstrates that cleavage-stage FISH poorly predicts aneuploidy in embryos that develop to morphologically normal blastocysts. Mol Hum Reprod 16: 590–600, 2010. Nowaczyk MJM, Zeesman S, Kam A , Taylor SAM, Carter RF, Whelan DT. Boy with 47,XXY, del(15) (q11.2q13) karyotype and Prader-Willi syndrome: a new case and review of the literature. Am J Med Genet 125A : 73–76, 2004. Nowicki ST, Tassone F, Ono MY, et al. The Prader-Willi phenotype of fragile X syndrome. J Dev Behav Pediatr 28: 133–138, 2007. Nowinski GP, Van Dyke DL, Tilley BC, et al. The frequency of aneuploidy in cultured lymphocytes is correlated with age and gender but not with reproductive history. Am J Hum Genet 46: 1101–1111, 1990. Nucaro AL, Falchi M, Pisano T, et al. Ring chromosome 14 mosaicism: an unusual case associated with developmental delay and epilepsy, characterized by genome array-CGH. Am J Med Genet 152A : 234–236, 2010. Nucaro AL, Meloni M, Pisano T, et al. Familial translocation t(3;10) (p26.3; p12.31) leading to trisomy 10p12.31→pter and monosomy 3p26.3→pter in seven members. Am J Med Genet 146A : 3242–3245, 2008. Nucaro AL, Rossino R , Pruna D, et al. Prenatal diagnosis of a mosaic supernumerary marker iso
(8p) (tetrasomy 8p): discordance between chorionic villi culture and amniotic fluid karyotypes. Prenat Diagn 26: 418–419, 2006. Ogasawara M, Aoki K , Okada S, Suzumori K . Embryonic karyotype of abortuses in relation to the number of previous miscarriages. Fertil Steril 73: 300–304, 2000. Ogata T, Matsuo M, Muroya K , Koyama Y, Fukutani K . 47,XXX male: a clinical and molecular study. Am J Med Genet 98: 353–356, 2001. Ogilvie CM, Yaron Y, Beaudet AL. Current controversies in prenatal diagnosis 3: for prenatal diagnosis, should we offer less or more than metaphase karyotyping? Prenat Diagn 29: 11–14, 2009. Ogur G, Van Assche E, Vegetti W, et al. Chromosomal segregation in spermatozoa of 14 Robertsonian translocation carriers. Mol Hum Reprod 12: 209–215, 2006. Ohye T, Inagaki H, Kogo H, et al. Paternal origin of the de novo constitutional t(11;22)(q23;q11). Eur J Hum Genet 18: 783–787, 2010. Okamoto N, Kubota T, Nakamura Y, et al. 22q13 Microduplication in two patients with common clinical manifestations: a recognizable syndrome? Am J Med Genet 143A : 2804–2809, 2007. Okun N, Summers AM, Hoffman B, et al. Prospective experience with integrated prenatal screening and first trimester combined screening for trisomy 21 in a large Canadian urban center. Prenat Diagn 28: 987–992, 2008. Olander E, Stamberg J, Steinberg L, Wulfsberg EA . Third Prader-Willi syndrome phenotype due to maternal uniparental disomy 15 with mosaic trisomy 15. Am J Med Genet 93: 215–218, 2000. Oliver-Bonet M, Benet J, Sun F, et al. Meiotic studies in two human reciprocal translocations and their association with spermatogenic failure. Hum Reprod 20: 683–688, 2005. Oliver-Bonet M, Navarro J, Codina-Pascual M, et al. From spermatocytes to sperm: meiotic behaviour of human male reciprocal translocations. Hum Reprod 19: 2515–2522, 2004. Olshan AF, Baird PA , Teschke K . Paternal occupational exposures and the risk of Down syndrome. Am J Hum Genet 44: 646–651, 1989. Ono T, Okuma M, Hamada T, Motohashi N, Moriyama K . A case of ring chromosome 18 syndrome treated with a combinedorthodonticprosthodontic approach. Cleft Palate Craniofac J 47: 201–210, 2010. Oostra BA , Willemsen R . FMR1: a gene with three faces. Biochim Biophys Acta 1790: 467–477, 2009.
Opheim KE, Brittingham A , Chapman D, Norwood TH. Balanced reciprocal translocation mosaicism: how frequent? Am J Med Genet 57: 601–604, 1995. Oracova E, Musilova P, Kopecna O, et al. Sperm and embryo analysis in a carrier of supernumerary inv dup(15) marker chromosome. J Androl 30: 233–239, 2009. Orellana C, Bernabeu J, Monfort S, et al. Duplication of the Williams-Beuren critical region: case report and further delineation of the phenotypic spectrum. J Med Genet 45: 187–189, 2008. Orellana C, Martínez F, Badía L, et al. Trisomy rescue by postzygotic unbalanced (X;14) translocation in a girl with dysmorphic features. Clin Genet 60: 206–211, 2001. Ortenberg J, Oddoux C, Craver R , et al. Sry gene expression in the ovotestes of XX true hermaphrodites. J Urol 167: 1828–1831, 2002. Ostrer H, Stamberg J, Perinchief P. Two chromosome aberrations in the child of a woman with systemic lupus erythematosus treated with azathioprine and prednisone. Am J Med Genet 17: 627–632, 1984. Otani T, Roche M, Mizuike M, Colls P, Escudero T, Munné S. Preimplantation genetic diagnosis significantly improves the pregnancy outcome of translocation carriers with a history of recurrent miscarriage and unsuccessful pregnancies. Reprod Biomed Online 13: 869–874, 2006. Otsuka S, Sakamoto Y, Siomi H, et al. Fragile X carrier screening and FMR1 allele distribution in the Japanese population. Brain Dev 32: 110–114, 2010. Otter M, Schrander-Stumpel CTRM, Curfs LMG. Triple X syndrome: a review of the literature. Eur J Hum Genet 18: 265–271, 2010. Ou Z, Berg JS, Yonath H, et al. Microduplications of 22q11.2 are frequently inherited and are associated with variable phenotypes. Genet Med 10: 267–277, 2008a. Ou Z, Martin DM, Bedoyan JK , et al. Branchiootorenal syndrome and oculoauriculovertebral spectrum features associated with duplication of SIX1, SIX6, and OTX2 resulting from a complex chromosomal rearrangement. Am J Med Genet 146A : 2480– 2489, 2008b. Ouldim K , Sbiti A , Natiq A , El-Kerch F, Cherkaoui S, Sefiani A . Unexpected fertility and paternal UPD 22. Fertil Steril 90: 2013 e13–15, 2008. Ozawa N, Maruyama T, Nagashima T, et al. Pregnancy outcomes of reciprocal translocation carriers who have a history of repeated pregnancy loss. Fertil Steril 90: 1301–1304, 2008. Ozen M, Hopwood VL, Pathak S. Ag-NOR studies in a human lymphocyte culture: are variants localized
REFERENCES
•
581
to specific chromosomes? Am J Med Genet 59: 225–228, 1995. Pagnamenta AT, Wing K , Sadighi Akha E, et al. A 15q13.3 microdeletion segregating with autism. Eur J Hum Genet 17: 687–692, 2009. Pai GS, Shields SM, Houser PM. Segregation of inverted chromosome 13 in families ascertained through liveborn recombinant offspring. Am J Med Genet 27: 127–133, 1987. Palermo GD, Colombero LT, Hariprashad JJ, Schlegel PN, Rosenwaks Z. Chromosome analysis of epididymal and testicular sperm in azoospermic patients undergoing ICSI. Hum Reprod 17: 570–575, 2002. Palka G, Calabrese G, Stuppia L, et al. A woman with an apparent non-mosaic 45,X delivered a 46,X,der(X) liveborn female. Clin Genet 45: 93–96, 1994. Panasiuk B, Danik J, Lurie IW, et al. Reciprocal chromosome translocations involving short arm of chromosome 9 as a risk factor of unfavorable pregnancy outcomes after meiotic malsegregation 2:2. Adv Med Sci 54: 203–210, 2009. Panasiuk B, Leśniewicz R , Spółczyńska A , et al. Translocation form of Wolf-Hirschhorn syndrome —assessment of recurrence rate probability. Adv Med Sci 52(suppl 1): 166–170, 2007. Panasiuk B, Ušinskiené R , Kostyk E, et al. Genetic counselling in carriers of reciprocal chromosomal translocations involving short arm of chromosome X . Ann Génét 47: 11–28, 2004. Pang MG, Hoegerman SF, Cuticchia AJ, et al. Detection of aneuploidy for chromosomes 4, 6, 7, 8, 9, 10, 11, 12, 13, 17, 18, 21, X and Y by fluorescence in-situ hybridization in spermatozoa from nine patients with oligoasthenoteratozoospermia undergoing intracytoplasmic sperm injection. Hum Reprod 14: 1266–1273, 1999. Pangalos C, Avramopoulos D, Blouin JL, et al. Understanding the mechanism(s) of mosaic trisomy 21 by using DNA polymorphism analysis. Am J Hum Genet 54: 473–481, 1994. Pangalos C, Théophile D, Sinet PM, et al. No significant effect of monosomy for distal 21q22.3 on the Down syndrome phenotype in “mirror” duplications of chromosome 21. Am J Hum Genet 51: 1240–1250, 1992a. Pangalos CG, Talbot CC, Lewis JG, et al. DNA polymorphism analysis in families with recurrence of free trisomy 21. Am J Hum Genet 51: 1015–1027, 1992b. Pankau R , Siebert R , Kautza M, et al. Familial Williams-Beuren syndrome showing varying
582
•
REFERENCES
clinical expression. Am J Med Genet 98: 324–329, 2001. Paoloni-Giacobino A , Dahoun SP, Sizonenko PC, Stalberg A , Chardonnens D, Campana A . A case of 45,X Turner syndrome with spontaneous ovulation proven by ultrasonography. Gynecol Endocrinol 14: 105–110, 2000a. Paoloni-Giacobino A , Kern I, Rumpler Y, Djlelati R , Morris MA , Dahoun SP. Familial t(6;21) (p21.1;p13) translocation associated with male-only sterility. Clin Genet 58: 324–328, 2000b. Paoloni-Giacobino A , Lespinasse J. Chromosome Y polysomy: a non-mosaic 49,XYYYY case. Clin Dysmorphol 16: 65–66, 2007. Papadakis V, Vlachopapadopoulou E, Van Syckle K , et al. Gonadal function in young patients successfully treated for Hodgkin disease. Med Pediatr Oncol 32: 366–372, 1999. Papanikolaou EG, Vernaeve V, Kolibianakis E, et al. Is chromosome analysis mandatory in the initial investigation of normovulatory women seeking infertility treatment? Hum Reprod 20: 2899–2903, 2005. Papenhausen P, Schwartz S, Risheg H, et al. UPD detection using homozygosity profiling with a SNP genotyping microarray. Am J Med Genet 155A : 757–768, 2011. Papoulidis I, Manolakos E, Siomou E, et al. A fetus with ring chromosome 21 characterized by aCGH shows no clinical findings after birth. Prenat Diagn 30: 586–588, 2010. Pareyson D, Testa D, Morbin M, et al. Does CMT1A homozygosity cause more severe disease with root hypertrophy and higher CSF proteins? Neurology 60: 1721–1722, 2003. Park JP, Moeschler JB, Hani VH, et al. Maternal disomy and Prader-Willi syndrome consistent with gamete complementation in a case of familial translocation (3;15) (p25;q11.2). Am J Med Genet 78: 134–139, 1998. Parker EA , Hovanes K , Germak J, Porter F, Merke DP. Maternal 21-hydroxylase deficiency and uniparental isodisomy of chromosome 6 and X results in a child with 21-hydroxylase deficiency and Klinefelter syndrome. Am J Med Genet 140: 2236–2240, 2006. Parma P, Radi O, Vidal V, et al. R-spondin1 is essential in sex determination, skin differentiation and malignancy. Nat Genet 38: 1304–1309, 2006. Parmar RC, Muranjan MN, Kotvaliwale S, Sharma S, Bharucha BA . Ring chromosome 12 with variable phenotypic features: clinical report and review of the literature. Am J Med Genet 117A : 275–277, 2003.
Parrish JE, Oostra BA , Verkerk AJMH, et al. Isolation of a GCC repeat showing expansion in FRAXF, a fragile site distal to FRAXA and FRAXE. Nat Genet 8: 229–235, 1994. Paskulin GA , Riegel M, Rosa RFM, Graziadio C, Zen PRG. Interstitial deletion of 7q31.32→q33 secondary to a paracentric inversion of a maternal chromosome 7. Eur J Med Genet 54: 181–185, 2011. Pasquini L, Tondi F, Pontello V, Filippeschi M. First trimester diagnosis of hypotriploidy (68,XX) with elevated fetal head to trunk volume ratio and small placental volume. Prenat Diagn 30: 589–590, 2010. Pasquino AM, Passeri F, Pucarelli I, Segni M, Municchi G. Spontaneous pubertal development in Turner’s syndrome. Italian Study Group for Turner’s Syndrome. J Clin Endocrinol Metab 82: 1810–1813, 1997. Patel ZM, Madon P. Interchange trisomy 22 in a live born resulting from 3:1 segregation in a t(15;22) (p12;q13) carrier mother. Indian J Pediatr 71: 1042, 2004. Patwardhan AJ, Brown WE, Bender BG, Linden MG, Eliez S, Reiss AL. Reduced size of the amygdala in individuals with 47,XXY and 47,XXX karyotypes. Am J Med Genet 114: 93–98, 2002. Pauli RM, Pagon RA , Hall JG. Trisomy 18 in sibs and maternal chromosome 9 variant. Birth Defects Orig Artic Ser 14: 297–301, 1978. Pavlidis K , McCauley E, Sybert VP. Psychosocial and sexual functioning in women with Turner syndrome. Clin Genet 47: 85–89, 1995. Paylor R , Glaser B, Mupo A , et al. Tbx1 haploinsufficiency is linked to behavioral disorders in mice and humans: implications for 22q11 deletion syndrome. Proc Natl Acad Sci USA 103: 7729–7734, 2006. Pazooki M, Lebbar A , Roubergues A , Baverel F, Letessier D, Dupont JM. Pure familial 6q21q22.1 duplication in two generations. Eur J Med Genet 50: 60–65, 2007. Pehlivan T, Pober BR , Brueckner M, et al. GATA4 haploinsufficiency in patients with interstitial deletion of chromosome region 8p23.1 and congenital heart disease. Am J Med Genet 83: 201–206, 1999. Peitsidis P, Manolakos E, Peitsidou A , et al. Pentasomy 49,XXXXY diagnosed in utero: case report and systematic review of antenatal findings. Fetal Diagn Ther 26: 1–5, 2009. Pelias MZ, Shaw MW. Torts of wrongful birth and wrongful life: a review. Am J Medi Genet 25: 71–80, 1986. Pellestor F. Analysis of meiotic segregation in a man heterozygous for a 13;15 Robertsonian
translocation and a review of the literature. Hum Genet 85: 49–54, 1990. Pellestor F, Anahory T, Hamamah S. Effect of maternal age on the frequency of cytogenetic abnormalities in human oocytes. Cytogenet Genome Res 111: 206–212, 2005. Pellestor F, Andréo B, Anahory T, Hamamah S. The occurrence of aneuploidy in human: lessons from the cytogenetic studies of human oocytes. Eur J Med Genet 49: 103–116, 2006. Pellestor F, Andréo B, Arnal F, Humeau C, Demaille J. Mechanisms of non-disjunction in human female meiosis: the co-existence of two modes of malsegregation evidenced by the karyotyping of 1397 in-vitro unfertilized oocytes. Hum Reprod 17: 2134–2145, 2002. Pellestor F, Andréo B, Arnal F, Humeau C, Demaille J. Maternal aging and chromosomal abnormalities: new data drawn from in vitro unfertilized human oocytes. Hum Genet 112: 195–203, 2003. Pellestor F, Imbert I, Andréo B, Lefort G. Study of the occurrence of interchromosomal effect in spermatozoa of chromosomal rearrangement carriers by fluorescence in-situ hybridization and primed in-situ labelling techniques. Hum Reprod 16: 1155–1164, 2001. Pellicer A , Rubio C, Vidal F, et al. In vitro fertilization plus preimplantation genetic diagnosis in patients with recurrent miscarriage: an analysis of chromosome abnormalities in human preimplantation embryos. Fertil Steril 71: 1033–1039, 1999. Peng HH, Chao AS, Wang TH, Chang YL, Chang SD. Prenatally diagnosed balanced chromosome rearrangements: eight years’ experience. J Reprod Med 51: 699–703, 2006. Penny LA , Dell’Aquila M, Jones MC, et al. Clinical and molecular characterization of patients with distal 11q deletions. Am J Hum Genet 56: 676–683, 1995. Penrose LS. The relative effects of paternal and maternal age in mongolism. J Genet 27: 219–224, 1933. Penrose LS. The relative aetiological importance of birth order and maternal age in mongolism. Proc Roy Soc B 115: 431–450, 1934. Penrose LS. Some notes on heredity counseling. Acta Genet Stat Med 6: 35–40, 1956. Pergament E. By 2020 . . . Prenat Diagn 30: 710–711, 2010. Pergament E, Confino E, Zhang JX , Roscetti L, Xien Chen P, Wellman D. Recurrent triploidy of maternal origin. Prenat Diagn 20: 561–563, 2000. Perrin A , Douet-Guilbert N, Le Bris MJ, et al. Segregation of chromosomes in sperm of a
REFERENCES
•
583
t(X;18)(q11;p11.1) carrier inherited from his mother: case report. Hum Reprod 23: 227–230, 2008. Perrin A , Douet-Guilbert N, Le Bris MJ, et al. Increased aneuploidy rates in spermatozoa of a male carrier of a trisomy 18 mosaicism. Int J Androl 32: 231–234, 2009. Perry J, Slater HR , Choo KHA . Centric fission— simple and complex mechanisms. Chromosome Res 12: 627–640, 2004. Perry J, White SM, Nouri S, et al. Unstable Robertsonian translocations der(13;15) (q10;q10): heritable chromosome fission without phenotypic effect in two kindreds. Am J Med Genet 136: 25–30, 2005. Perry MJ. Effects of environmental and occupational pesticide exposure on human sperm: a systematic review. Hum Reprod Update 14: 233–242, 2008. Persutte WH, Lenke RR . Failure of amniotic-fluid-cell growth: is it related to fetal aneuploidy? Lancet 345: 96–97, 1995. Pertile MD, Ryan JT, Kelley JL, Webber LM. Prenatal diagnosis of tetrasomy 9p. Bull Hum Genet Soc Australasia 9: 43, 1996. Pertl B, Kopp S, Kroisel PM, Tului L, Brambati B, Adinolfi M. Rapid detection of chromosome aneuploidies by quantitative fluorescence PCR: first application on 247 chorionic villus samples. J Med Genet 36: 300–303, 1999. Petersen MB, Adelsberger PA , Schinzel AA , Binkert F, Hinkel GK , Antonarakis SE. Down syndrome due to de novo Robertsonian translocation t(14q;21q): DNA polymorphism analysis suggests that the origin of the extra 21q is maternal. Am J Hum Genet 49: 529–536, 1991. Petignat P, Billieux MH, Blouin JL, Dahoun S, Vassilakos P. Is genetic analysis useful in the routine management of hydatidiform mole? Hum Reprod 18: 243–249, 2003. Petit F, Holder-Espinasse M, Duban-Bedu B, et al. Trisomy 7 mosaicism prenatally misdiagnosed and maternal uniparental disomy in a child with pigmentary mosaicism and Russell-Silver syndrome. Clin Genet in press (published online January 2011). Petković I, de Capoa A , Giancotti P, Barišić I. Unusual segregation of t(11;22) resulting from crossingover followed by 3:1 disjunction at meiosis I. Clin Genet 50: 515–519, 1996. Petracchi F, Igarzabal L, Crespo ML, Gadow E. Trisomy 16 detected by first trimester screening. Prenat Diagn 29: 1175–1176, 2009. Petrij F, Dauwerse HG, Blough RI, et al. Diagnostic analysis of the Rubinstein-Taybi syndrome: five cosmids should be used for microdeletion
584
•
REFERENCES
detection and low number of protein truncating mutations. J Med Genet 37: 168–176, 2000a. Petrij F, Dorsman JC, Dauwerse HG, et al. RubinsteinTaybi syndrome caused by a De Novo reciprocal translocation t(2;16)(q36.3;p13.3). Am J Med Genet 92: 47–52, 2000b. Petrovic V. A new variant of chromosome 3 with unusual staining properties. J Med Genet 25: 781–782, 1988. Pettenati MJ, Rao N, Johnson C, et al. Molecular cytogenetic analysis of a familial 8p23.1 deletion associated with minimal dysmorphic features, seizures, and mild mental retardation. Hum Genet 89: 602–606, 1992. Pettenati MJ, Rao PN, Phelan MC, et al. Paracentric inversions in humans: a review of 446 paracentric inversions with presentation of 120 new cases. Am J Med Genet 55: 171–187, 1995. Pettenati MJ, Rao PN, Weaver RG, Thomas IT, McMahan MR . Inversion (X)(p11.4q22) associated with Norrie disease in a four generation family. Am J Med Genet 45: 577–580, 1993. Pettenati MJ, Sweatt AJ, Lantz P, et al. The human cornea has a high incidence of acquired chromosome abnormalities. Hum Genet 101: 26–29, 1997. Pettenati MJ, Von Kap-Herr C, Jackle B, et al. Rapid interphase analysis for prenatal diagnosis of translocation carriers using subtelomeric probes. Prenat Diagn 22: 193–197, 2002. Pettenati MJ, Wheeler M, Bartlett DJ, et al. 45,X/47,XYY mosaicism: clinical discrepancy between prenatally and postnatally diagnosed cases. Am J Med Genet 39: 42–47, 1991. Pfeiffer RA , Loidl J. Mirror image duplications of chromosome 21. Three new cases and discussion of the mechanisms of origin. Hum Genet 62: 361–363, 1982. Pflueger S, Golden J, Troiano R , Gasparini R , Marini T. Fission of familial (13;22) Robertsonian translocation resulting in fetal mosaicism. Am J Hum Genet 49: 283, 1991. Phelan MC. Additional studies warranted to confirm monosomy 21. Prenat Diagn 22: 160–161, 2002. Phelan MC. Deletion 22q13.3 syndrome. Orphanet J Rare Dis 3: 14, 2008. Phelan MC, Crawford EC, Bealer DM. Mental retardation in South Carolina III. Chromosome aberrations. Proc Greenwood Genet Center 15: 45–60, 1996. Phelan MC, Rogers RC, Clarkson KB, et al. Albright hereditary osteodystrophy and del(2) (q37.3) in four unrelated individuals. Am J Med Genet 58: 1–7, 1995.
Phelan MC, Rogers RC, Stevenson RE. Multiple, compound, and complex chromosome rearrangements. Proc Greenwood Genet Center 9: 19–37, 1990. Phelan MC, Stevenson RE, Anderson EV, Jr. Recombinant chromosome 9 possibly derived from breakage and reunion of sister chromatids within a paracentric inversion loop. Am J Med Genet 46: 304–308, 1993. Philipp T, Philipp K , Reiner A , Beer F, Kalousek DK . Embryoscopic and cytogenetic analysis of 233 missed abortions: factors involved in the pathogenesis of developmental defects of early failed pregnancies. Hum Reprod 18: 1724–1732, 2003. Phillips OP, Tharapel AT, Lerner JL, Park VM, Wachtel SS, Shulman LP. Risk of fetal mosaicism when placental mosaicism is diagnosed by chorionic villus sampling. Am J Obstet Gynecol 174: 850–855, 1996. Piccini I, Ballarati L, Bassi C, et al. The structure of duplications on human acrocentric chromosome short arms derived by the analysis of 15p. Hum Genet 108: 467–477, 2001. Pickering DL, Eudy JD, Olney AH, et al. Array-based comparative genomic hybridization analysis of 1176 consecutive clinical genetics investigations. Genet Med 10: 262–266, 2008. Pietrzak J, Mrasek K , Obersztyn E, et al. Molecular cytogenetic characterization of eight small supernumerary marker chromosomes originating from chromosomes 2, 4, 8, 18, and 21 in three patients. J Appl Genet 48: 167–175, 2007. Pinho MJ, Neves R , Costa P, et al. Unique t(Y;1) (q12;q12) reciprocal translocation with loss of the heterochromatic region of chromosome 1 in a male with azoospermia due to meiotic arrest: a case report. Hum Reprod 20: 689–696, 2005. Pinto Leite R , Pinto M. Prenatal detection of an inverted X chromosome in a male fetus. Prenat Diagn 21: 233, 2001. Pipiras E, Dupont C, Chantot-Bastaraud S, et al. Structural chromosomal mosaicism and prenatal diagnosis. Prenat Diagn 24: 101–103, 2004. Pittalis MC, Santarini L, Bovicelli L. Prenatal diagnosis of a heterochromatic 18p+ heteromorphism. Prenat Diagn 14: 72–73, 1994. Pivnick EK , Wilroy RS, Summitt JB, Tucker B, Herrod HG, Tharapel AT. Adjacent-2 disjunction of a maternal t(9;22) leading to duplication 9pter→q22 and deficiency of 22pter→q11.2. Am J Med Genet 37: 92–96, 1990. Plaja A , Mediano C, Cano L, et al. Prenatal diagnosis of a rare chromosomal instability syndrome: variegated aneuploidy related to premature
centromere division (PCD). Am J Med Genet 117A : 85–86, 2003. Pollin TI, Dobyns WB, Crowe CA , Ledbetter DH, Bailey-Wilson JE, Smith AC. Risk of abnormal pregnancy outcome in carriers of balanced reciprocal translocations involving the MillerDieker syndrome (MDS) critical region in chromosome 17p13.3. Am J Med Genet 85: 369–375, 1999. Portnoï MF. Microduplication 22q11. 2: a new chromosomal syndrome. Eur J Med Genet 52:88–93, 2009. Potocki L, Bi W, Treadwell-Deering D, et al. Characterization of Potocki-Lupski syndrome (dup(17)(p11.2p11.2)) and delineation of a dosage-sensitive critical interval that can convey an autism phenotype. Am J Hum Genet 80: 633–649, 2007. Potocki L, Chen KS, Park SS, et al. Molecular mechanism for duplication 17p11.2- the homologous recombination reciprocal of the Smith-Magenis microdeletion. Nat Genet 24: 84–87, 2000a. Potocki L, Glaze D, Tan DX , et al. Circadian rhythm abnormalities of melatonin in Smith-Magenis syndrome. J Med Genet 37: 428–433, 2000b. Potocki L, Shaffer LG. Interstitial deletion of 11(p11.2p12): a newly described contiguous gene deletion syndrome involving the gene for hereditary multiple exostoses (EXT2). Am J Med Genet 62: 319–325, 1996. Póvoa A , Ramalho C, Torgal A , et al. Positive biochemical screening for trisomy 18: on the path of trisomy 9. Prenat Diagn 28: 162–164, 2008. Pradhan M, Dalal A , Khan F, Agrawal S. Fertility in men with Down syndrome: a case report. Fertil Steril 86: 1765 e1–3, 2006. Prasad C, Wagstaff J. Genotype and phenotype in Angelman syndrome caused by paternal UPD 15. Am J Med Genet 70: 328–329, 1997. Prasad SE, Howley S, Murphy KC. Candidate genes and the behavioral phenotype in 22q11.2 deletion syndrome. Dev Disabil Res Rev 14: 26–34, 2008. Preis S, Majewski F. Monozygotic twins concordant for Rubinstein-Taybi syndrome: changing phenotype during infancy. Clin Genet 48: 72–75, 1995. Price HA , Roberts SH, Laurence KM. Homozygous paracentric inversion 12 in a mentally retarded boy: a case report and review of the literature. Hum Genet 75: 101–108, 1987. Price N, Bahra M, Griffin D, Hanna G, Stock A . Cornelia de Lange Syndrome in association with a balanced reciprocal translocation involving
REFERENCES
•
585
chromosomes 3 and 5. Prenat Diagn 25: 602–603, 2005. Priola V, De Vivo A , Imbesi G, Azzerboni A , Triolo O. Trisomy 9 associated with maternal serum screening results positive for trisomy 18. Case report and review of the literature. Prenat Diagn 27: 1167–1169, 2007. Pritchard MA , Kola I. The “gene dosage effect” hypothesis versus the “amplified developmental instability” hypothesis in Down syndrome. J Neural Transm Suppl 57: 293–303, 1999. Prontera P, Buldrini B, Aiello V, et al. Trisomy 15 mosaicism owing to familial reciprocal translocation t(1;15): implication for prenatal diagnosis. Prenat Diagn 26: 571–576, 2006. Puck MH. Some considerations bearing on the doctrine of self-fulfilling prophecy in sex chromosome aneuploidy. Am J Med Genet 9: 129–137, 1981. Pujol A , Benet J, Staessen C, et al. The importance of aneuploidy screening in reciprocal translocation carriers. Reproduction 131: 1025–1035, 2006. Pujol A , Durban M, Benet J, et al. Multiple aneuploidies in the oocytes of balanced translocation carriers: a preimplantation genetic diagnosis study using first polar body. Reproduction 126: 701–711, 2003. Punnett A , Teshima I, Heon E, et al. Unique insertional translocation in a childhood Wilms’ tumor survivor detected when his daughter developed bilateral retinoblastoma. Am J Med Genet 120A : 105–109, 2003. Purandare SM, Lee J, Hassed S, et al. Ring chromosome 9 [r(9)(p24q34)]: a report of two cases. Am J Med Genet 138A : 229–235, 2005. Purvis-Smith SG, Saville T, Manass S, et al. Uniparental disomy 15 resulting from “correction” of an initial trisomy 15. Am J Hum Genet 50: 1348–1350, 1992. Putzova M, Soldatova I, Pecnova L, et al. QF-PCRbased prenatal detection of common aneuploidies in the Czech population: five years of experience. Eur J Med Genet 51: 209–218, 2008. Puvabanditsin S, Garrow E, Razi S, et al. A Y/15 translocation in a 45,X male with Prader-Willi syndrome. Genet Couns 18: 417–421, 2007. Qian J, Cheng Q, Murdoch S, et al. The genetics of recurrent hydatidiform moles in China: Correlations between NLRP7 mutations, molar genotypes, and reproductive outcomes. Mol Hum Reprod in press (published online April 2011). Qiao Y, Liu X , Harvard C, et al. Autism-associated familial microdeletion of Xp11.22. Clin Genet 74: 134–144, 2008.
586
•
REFERENCES
Quack B, Speed RM, Luciani JM, Noel B, Guichaoua M, Chandley AC. Meiotic analysis of two human reciprocal X-autosome translocations. Cytogenet Cell Genet 48: 43–47, 1988. Quan F, Janas J, Toth-Fejel S, Johnson DB, Wolford JK , Popovich BW. Uniparental disomy of the entire X chromosome in a female with Duchenne muscular dystrophy. Am J Hum Genet 60: 160–165, 1997. Quan F, Zonana J, Gunter K , Peterson KL, Magenis RE, Popovich BW. An atypical case of fragile X syndrome caused by a deletion that includes the FMR1 gene. Am J Hum Genet 56: 1042–1051, 1995. Queipo G, Zenteno JC, Peña R , et al. Molecular analysis in true hermaphroditism: demonstration of low-level hidden mosaicism for Y-derived sequences in 46,XX cases. Hum Genet 111: 278–283, 2002. Quélin C, Bendavid C, Dubourg C, et al. Twelve new patients with 13q deletion syndrome: genotypephenotype analyses in progress. Eur J Med Genet 52: 41–46, 2009. Quenum-Miraillet G, Malan V, Martinovic J, et al. Prenatal diagnosis of a ring chromosome 14 in a fetus with a severe skeletal dysplasia. Prenat Diagn 28: 69–71, 2008. Queralt R , Madrigal I, Vallecillos MA , et al. Atypical XX male with the SRY gene located at the long arm of chromosome 1 and a 1qter microdeletion. Am J Med Genet 146A : 1335–1340, 2008. Quigley DI, McDonald MT, Krishnamuthy V, et al. Triploid mosaicism in a 45,X/69,XXY infant. Am J Med Genet 138A : 171–174, 2005. Quilter CR , Svennevik EC, Serhal P, et al. Cytogenetic and Y chromosome microdeletion screening of a random group of infertile males. Fertil Steril 79: 301–307, 2003. Qumsiyeh MB, Kim KR , Ahmed MN, Bradford W. Cytogenetics and mechanisms of spontaneous abortions: increased apoptosis and decreased cell proliferation in chromosomally abnormal villi. Cytogenet Cell Genet 88: 230–235, 2000. Qumsiyeh MB, Rafi SK , Sarri C, et al. Double supernumerary isodicentric chromosomes derived from 15 resulting in partial hexasomy. Am J Med Genet 116A : 356–359, 2003. Raca G, Artzer A , Thorson L, et al. Array-based comparative genomic hybridization (aCGH) in the genetic evaluation of stillbirth. Am J Med Genet 149A : 2437–2443, 2009. Rajangam S, Michaelis RC, Velagaleti GVN, et al. Down syndrome with biparental inheritance of der(14q21q) and maternally derived trisomy 21: confirmation by fluorescent in situ hybridization
and microsatellite polymorphism analysis. Am J Med Genet 70: 43–47, 1997. Rajcan-Separovic E, Qiao Y, Tyson C, et al. Genomic changes detected by array CGH in human embryos with developmental defects. Mol Hum Reprod 16: 125–134, 2010. Rajcan-Separovic E, Robinson WP, Stephenson M, et al. Recurrent trisomy 15 in a female carrier of der(15)t(Y;15)(q12;p13). Am J Med Genet 99: 320–324, 2001. Ralph A , Scott F, Tiernan C, et al. Maternal uniparental isodisomy for chromosome 14 detected prenatally. Prenat Diagn 19: 681–684, 1999. Ramos-Corpas D, Santiago JC, Montoya F. Ultrasonographic evaluation of fetal nasal bone in a low-risk population at 11–13 + 6 gestational weeks. Prenat Diagn 26: 112–117, 2006. Ramsay L, Howe DT, Wellesley D. Parental attitude to participating in long-term follow-up studies of their children’s health after in utero diagnosis of abnormalities. Prenat Diagn 29: 207–212, 2009. Ratcliffe S. Long-term outcome in children of sex chromosome abnormalities. Arch Dis Child 80: 192–195, 1999. Ratcliffe SG, Butler GE, Jones M. Edinburgh study of growth and development of children with sex chromosome abnormalities. IV. Birth Defects Orig Artic Ser 26: 1–44, 1990. Rauch A , Beese M, Mayatepek E, et al. A novel 5q35.3 subtelomeric deletion syndrome. Am J Med Genet 121A : 1–8, 2003. Rauen KA , Bitts SM, Li L, Golabi M, Cotter PD. Tandem duplication mosaicism: characterization of a mosaic dup(5q) and review. Clin Genet 60: 366–370, 2001. Rauen KA , Golabi M, Cotter PD. Fertility in a female with mosaic trisomy 8. Fertil Steril 79: 206–208, 2003. Ravel C, Berthaut I, Bresson JL, Siffroi JP. Prevalence of chromosomal abnormalities in phenotypically normal and fertile adult males: large-scale survey of over 10,000 sperm donor karyotypes. Hum Reprod 21: 1484–1489, 2006a. Ravel C, Chantot-Bastaraud S, Siffroi JP, Escalier D, Antoine JM, Mandelbaum J. Tail stump syndrome associated with chromosomal translocation in two brothers attempting intracytoplasmic sperm injection. Fertil Steril 86: 719 e1–7, 2006b. Ravnan JB, Tepperberg JH, Papenhausen P, et al. Subtelomere FISH analysis of 11 688 cases: an evaluation of the frequency and pattern of subtelomere rearrangements in individuals with developmental disabilities. J Med Genet 43: 478–489, 2006.
Ray JG, Meier C, Vermeulen MJ, Cole DEC, Wyatt PR . Prevalence of trisomy 21 following folic acid food fortification. Am J Med Genet 120A : 309–313, 2003. Raziel A , Friedler S, Schachter M, Kasterstein E, Strassburger D, Ron-El R . Increased frequency of female partner chromosomal abnormalities in patients with high-order implantation failure after in vitro fertilization. Fertil Steril 78: 515–519, 2002. Reches A , Malcov M, Ben-Yosef D, Azem F, Amit A , Yaron Y. Preimplantation genetic diagnosis for fragile X syndrome: is there increased transmission of abnormal FMR1 alleles among female heterozygotes? Prenat Diagn 29: 57–61, 2009. Redaelli S, Sala E, Roncaglia N, et al. Severe intrauterine growth restriction and trisomy 15 confined placental mosaicism: a case report and review of literature. Prenat Diagn 25: 140–147, 2005. Reddy KS. Double trisomy in spontaneous abortions. Hum Genet 101: 339–345, 1997. Reddy KS, Huang B. Prenatal management of mosaic tetrasomy 5p. Prenat Diagn 23: 82–85, 2003. Reddy KS, Mak L. Mosaic unbalanced structural abnormalities confirmed using FISH on buccal mucosal cells. Ann Génét 44: 37–40, 2001. Reddy KS, Sulcova V. The mobile nature of acrocentric elements illustrated by three unusual chromosome variants. Hum Genet 102: 653–662, 1998a. Reddy KS, Sulcova V. Pathogenetics of 45,X/46,XY gonadal mosaicism. Cytogenet Cell Genet 82: 52–57, 1998b. Reddy KS, Sulcova V, Siassi B. Two sibs with Wolf-Hirschhorn and DiGeorge deletions resulting from an unbalanced chromosome rearrangement, 45,XX/XY, der(4)t(4;22) (p16.3;q11.2) mat,-22. J Med Genet 33: 852–855, 1996. Reddy KS, Sulcova V, Young H, Blancato JK , Haddad BR . De novo mosaic add(3) characterized to be trisomy 14q31-qter using spectral karyotyping and subtelomeric probes. Am J Med Genet 82: 318–321, 1999. Redon R , Ishikawa S, Fitch KR , et al. Global variation in copy number in the human genome. Nature 444: 444–454, 2006. Reichenbach H, Holland H, Dalitz E, et al. De novo complete trisomy 5p: clinical report and FISH studies. Am J Med Genet 85: 447–451, 1999. Reid R , Sepulveda W, Kyle PM, Davies G. Amniotic fluid culture failure: clinical significance and association with aneuploidy. Obstet Gynecol 87: 588–592, 1996.
REFERENCES
•
587
Reinehr T, Jauch A, Zoll B, Engel U, Bartels I, Andler W. Short stature in a mother and daughter caused by familial der(X)t(X;X)(p22.1–3;q26). Am J Med Genet 102: 81–85, 2001. Reish O, Wolach B, Amiel A , Kedar I, Dolfin T, Fejgin M. Dilemma of trisomy 20 mosaicism detected prenatally: is it an innocent finding? Am J Med Genet 77: 72–75, 1998. Reiss AL, Freund L, Plotnick L, et al. The effects of X monosomy on brain development: monozygotic twins discordant for Turner’s syndrome. Ann Neurol 34: 95–107, 1993. Rentmeester CA . Value neutrality in genetic counseling: an unattained ideal. Med Health Care Philos 4: 47–51, 2001. Rethoré MO, Blois MC, Peeters M, Popowski P, Pangalos C, Lejeune J. Pure partial trisomy of the short arm of chromosome 5. Hum Genet 82: 296–298, 1989. Reyniers E, Vits L, De Boulle K , et al. The full mutation in the FMR-1 gene of male fragile X patients is absent in their sperm. Nat Genet 4: 143–146, 1993. Ribeiro Noce T, de Pina-Neto JM, Happle R . Phylloid pattern of pigmentary disturbance in a case of complex mosaicism. Am J Med Genet 98: 145–147, 2001. Ricard-Mousnier B, N’Guyen S, Dubas F, Pouplard F, Guichet A . Ring chromosome 17 epilepsy may resemble that of ring chromosome 20 syndrome. Epileptic Disord 9: 327–331, 2007. Riccaboni A, Lalatta F, Caliari I, Bonetti S, Somigliana E, Ragni G. Genetic screening in 2,710 infertile candidate couples for assisted reproductive techniques: results of application of Italian guidelines for the appropriate use of genetic tests. Fertil Steril 89: 800–808, 2008. Rickman L, Fiegler H, Shaw-Smith C, et al. Prenatal detection of unbalanced chromosomal rearrangements by array CGH. J Med Genet 43: 353–361, 2006. Riegel M, Baumer A , Jamar M, et al. Submicroscopic terminal deletions and duplications in retarded patients with unclassified malformation syndromes. Hum Genet 109: 286–294, 2001. Riegel M, Wisser J, Baumer A , Schinzel A . Postzygotic isochromosome formation as a cause for falsenegative results from chorionic villus chromosome examinations. Prenat Diagn 26: 221–225, 2006. Rieubland C, Francis D, Houben L, Corrie S, Bankier A , White SM. Two cases of trisomy 16 mosaicism ascertained postnatally. Am J Med Genet 149A : 1523–1528, 2009. Rigola MA , Carrera M, Ribas I, Egozcue J, Miro R , Fuster C. A comparative genomic hybridization
588
•
REFERENCES
study in a 46,XX male. Fertil Steril 78: 186–188, 2002. Riley D, Wiznitzer M, Schwartz S, Zinn AB. A 13-yearold boy with cognitive impairment, retinoblastoma, and Wilson disease. Neurology 57: 141–143, 2001. Riley M, Halliday JL. “Birth defects in Victoria 1999–2000.” Perinatal data collection unit, Department of Human Services, State Government of Victoria, Australia, 2002. Rio M, Malan V, Boissel S, et al. Familial interstitial Xq27.3q28 duplication encompassing the FMR1 gene but not the MECP2 gene causes a new syndromic mental retardation condition. Eur J Hum Genet 18: 285–290, 2010. Rio M, Ozilou C, Cormier-Daire V, et al. Partial maternal heterodisomy of chromosome 17q25 in a case of severe mental retardation. Hum Genet 108: 511–515, 2001. Rivas F, García-Esquivel L, Rivera H, Jimenez ME, Gonzalez RM, Cantú JM. Inv(4)(p16q21). A five-generation pedigree with 24 carriers and no recombinants. Clin Genet 31: 97–101, 1987. Rivera H, Cantú JM. Centric fission consequences in man. Ann Génét 29: 223–225, 1986. Rivera H, Vásquez AI, Perea FJ. Centromere-telomere (12;8p) fusion, telomeric 12q translocation, and i(12p) trisomy. Clin Genet 55: 122–126, 1999. Rizzolio F, Bione S, Sala C, et al. Chromosomal rearrangements in Xq and premature ovarian failure: mapping of 25 new cases and review of the literature. Hum Reprod 21: 1477–1483, 2006. Rizzolio F, Pramparo T, Sala C, et al. Epigenetic analysis of the critical region I for premature ovarian failure: demonstration of a highly heterochromatic domain on the long arm of the mammalian X chromosome. J Med Genet 46: 585–592, 2009. Robbins WA , Vine MF, Truong KY, Everson RB. Use of fluorescence in situ hybridization (FISH) to assess effects of smoking, caffeine, and alcohol on aneuploidy load in sperm of healthy men. Environ Mol Mutagen 30: 175–183, 1997. Roberson EDO, Squibb Wohler E, Hoover-Fong , et al. Genomic analysis of partial 21q monosomies with variable phenotypes. Eur J Hum Genet 19: 235–238, 2011. Roberts E, Dunlop J, Davis GS, Churchill D, Davison EV. A further case of confined placental mosaicism for trisomy 2 associated with adverse pregnancy outcome. Prenat Diagn 23: 564–565, 2003. Roberts P, Williams J, Sills MA . A case of two inversion (10) recombinants in a family. J Med Genet 26: 461–464, 1989.
Roberts SE, Dennis NR , Browne CE, et al. Characterisation of interstitial duplications and triplications of chromosome 15q11-q13. Hum Genet 110: 227–234, 2002. Roberts SH, Hughes HE, Davies SJ, Meredith AL. Bilateral split hand and split foot malformation in a boy with a de novo interstitial deletion of 7q21.3. J Med Genet 28: 479–481, 1991. Robertson S, Savulescu J. Is there a case in favour of predictive genetic testing in young children? Bioethics 15: 26–49, 2001. Robinson A , Bender BG, Borelli JB, Puck MH, Salbenblatt JA , Winter JSD. Sex chromosomal aneuploidy: prospective and longitudinal studies. Birth Defects Orig Artic Ser 22: 23–71, 1986. Robinson A , Bender BG, Linden MG, Salbenblatt JA . Sex chromosome aneuploidy: the Denver Prospective Study. Birth Defects Orig Artic Ser 26: 59–115, 1990. Robinson DO, Jacobs PA . The origin of the extra Y chromosome in males with a 47,XYY karyotype. Hum Mol Genet 8: 2205–2209, 1999. Robinson WP, Barrett IJ, Bernard L, et al. Meiotic origin of trisomy in confined placental mosaicism is correlated with presence of fetal uniparental disomy, high levels of trisomy in trophoblast, and increased risk of fetal intrauterine growth restriction. Am J Hum Genet 60: 917–927, 1997. Robinson WP, Bernasconi F, Basaran S, et al. A somatic origin of homologous Robertsonian translocations and isochromosomes. Am J Hum Genet 54: 290–302, 1994. Robinson WP, Bernasconi F, Lau A , McFadden DE. Frequency of meiotic trisomy depends on involved chromosome and mode of ascertainment. Am J Med Genet 84: 34–42, 1999. Robinson WP, Binkert F, Bernasconi F, LordaSánchez I, Werder EA , Schinzel AA . Molecular studies of chromosomal mosaicism: relative frequency of chromosome gain or loss and possible role of cell selection. Am J Hum Genet 56: 444–451, 1995. Robinson WP, Binkert F, Giné R , et al. Clinical and molecular analysis of five inv dup(15) patients. Eur J Hum Genet 1: 37–50, 1993. Robinson WP, Christian SL, Kuchinka BD, et al. Somatic segregation errors predominantly contribute to the gain or loss of a paternal chromosome leading to uniparental disomy for chromosome 15. Clin Genet 57: 349–358, 2000. Robinson WP, Kuchinka BD, Bernasconi F, et al. Maternal meiosis I non-disjunction of chromosome 15: dependence of the maternal age effect on level of recombination. Hum Mol Genet 7: 1011–1019, 1998.
Robinson WP, McFadden DE, Barrett IJ, et al. Origin of amnion and implications for evaluation of the fetal genotype in cases of mosaicism. Prenat Diagn 22: 1076–1085, 2002. Robinson WP, McFadden DE, Stephenson MD. The origin of abnormalities in recurrent aneuploidy/ polyploidy. Am J Hum Genet 69: 1245–1254, 2001. Robinson WP, McGillivray B, Friedman JM. Pregnancy and postnatal outcome of mosaic isochromosome 20q. Prenat Diagn 27: 143–145, 2007. Robinson WP, McGillivray B, Lewis ME, Arbour L, Barrett I, Kalousek DK . Prenatally detected trisomy 20 mosaicism. Prenat Diagn 25: 239–244, 2005. Robinson WP, Peñaherrera MS, Jiang R , et al. Assessing the role of placental trisomy in preeclampsia and intrauterine growth restriction. Prenat Diagn 30: 1–8, 2010. Rodeck C, Ogilvie C. NICE goes off the rails. Prenat Diagn 29: 730–731, 2009. Rodrigo L, Peinado V, Mateu E, et al. Impact of different patterns of sperm chromosomal abnormalities on the chromosomal constitution of preimplantation embryos. Fertil Steril 94: 1380–1386, 2010. Rodríguez L, Diego-Alvarez D, Lorda-Sánchez I, et al. A small and active ring X chromosome in a female with features of Kabuki syndrome. Am J Med Genet 146A : 2816–2821, 2008. Rodriguez-Revenga L, Badenas C, Madrigal I, et al. 46,XY,18q+/46,XY,18q- mosaicism in a fragile X prenatal diagnosis. Prenat Diagn 25: 448–450, 2005. Rodriguez-Revenga L, Madrigal I, Pagonabarraga J, et al. Penetrance of FMR1 premutation associated pathologies in fragile X syndrome families. Eur J Hum Genet 17: 1359–1362, 2009. Rogers JC, Harris DJ, Pasztor LM. Reciprocal translocation 4;11 with both adjacent-1 segregants viable within a family. Clin Genet 51: 250–256, 1997. Rogers JG, Voullaire L, Gold H. Monozygotic twins discordant for trisomy 21. Am J Med Genet 11: 143–146, 1982. Rohr J, Allen EG, Charen K , et al. Anti-Mullerian hormone indicates early ovarian decline in fragile X mental retardation (FMR1) premutation carriers: a preliminary study. Hum Reprod 23: 1220–1225, 2008. Romain DR , Columbano-Green LM, Whyte S, et al. Familial paracentric inversion of 1p. Am J Med Genet 14: 629–634, 1983. Romain DR , Smythe RH, Columbano-Green LM, Chapman CJ, Sullivan J. Three unrelated cases of
REFERENCES
•
589
accessory bisatellited microchromosomes in New Zealand. NZ Med J 94: 90–91, 1981. Romain DR , Whyte S, Callen DF, Eyre HJ. A rare heteromorphism of chromosome 20 and reproductive loss. J Med Genet 28: 477–478, 1991. Romans SM, Stefanatos G, Roeltgen DP, Kushner H, Ross JL. Transition to young adulthood in Ullrich-Turner syndrome: neurodevelopmental changes. Am J Med Genet 79: 140–147, 1998. Ronan A , Fagan K , Christie L, Conroy J, Nowak NJ, Turner G. Familial 4.3 Mb duplication of 21q22 sheds new light on the Down syndrome critical region. J Med Genet 44: 448–451, 2007. Ron-El R , Strassburger D, Gelman-Kohan S, Friedler S, Raziel A , Appelman Z. A 47,XXY fetus conceived after ICSI of spermatozoa from a patient with non-mosaic Klinefelter’s syndrome: case report. Hum Reprod 15: 1804–1806, 2000. Rooryck C, VuPhi Y, Souakri N, et al. Characterization of a de novo balanced translocation t(9;18) (p23;q12.2) in a patient with oculoauriculovertebral spectrum. Eur J Med Genet 53: 104–107, 2010. Roos L, Jønch AE, Kjaergaard S, et al. A new microduplication syndrome encompassing the region of the Miller-Dieker (17p13 deletion) syndrome. J Med Genet 46: 703–710, 2009. Röpke A , Kalinski T, Mohnike K , et al. Distribution of sex chromosomes in dysgenetic gonads of mixed type. Cytogenet Genome Res 116: 146–151, 2007. Rose CSP, Patel P, Reardon W, Malcolm S, Winter RM. The TWIST gene, although not disrupted in Saethre-Chotzen patients with apparently balanced translocations of 7p21, is mutated in familial and sporadic cases. Hum Mol Genet 6: 1369–1373, 1997. Rosenberg MJ, Killoran C, Dziadzio L, et al. Scanning for telomeric deletions and duplications and uniparental disomy using genetic markers in 120 children with malformations. Hum Genet 109: 311–318, 2001. Rosenbusch B. Co-existence of a diploid hydatidiform mole and a normal fetus: mysterious events at the pronuclear stage. Hum Reprod 23: 2876–2877, 2008. Rosenbusch B, Schneider M, Gläser B, Brucker C. Cytogenetic analysis of giant oocytes and zygotes to assess their relevance for the development of digynic triploidy. Hum Reprod 17: 2388–2393, 2002. Rosenfeld JA , Ballif BC, Lucas A , et al. Small deletions of SATB2 cause some of the clinical features of the 2q33.1 microdeletion syndrome. PLoS One 4: e6568, 2009.
590
•
REFERENCES
Rosenfeld JA , Ballif BC, Torchia BS. Copy number variations associated with autism spectrum disorders contribute to a spectrum of neurodevelopmental disorders. Genet Med 12: 694–702, 2010a. Rosenfeld JA , Coppinger J, Bejjani B, et al. Speech delays and behavioral problems are the predominant features in individuals with developmental delays and 16p11.2 microdeletions and microduplications. J Neurodevel Disord 2: 26–38, 2010b. Rosenmann A , Wahrman J, Richler C, Voss R , Persitz A , Goldman B. Meiotic association between the XY chromosomes and unpaired autosomal elements as a cause of human male sterility. Cytogenet Cell Genet 39: 19–29, 1985. Rosias PR , Sijstermans JM, Theunissen PM, et al. Phenotypic variability of the cat eye syndrome. Case report and review of the literature. Genet Couns 12: 273–282, 2001. Ross JA , Jurkovic D, Nicolaides K . Coelocentesis: a study of short-term safety. Prenat Diagn 17: 913–917, 1997. Ross JL, Stefanatos GA , Kushner H, Zinn A , Bondy C, Roeltgen D. Persistent cognitive deficits in adult women with Turner syndrome. Neurology 58: 218–225, 2002. Rossi E, Messa J, Zuffardi O. Ring syndrome: still true? J Med Genet 45: 766–768, 2008a. Rossi E, Riegel M, Messa J, et al. Duplications in addition to terminal deletions are present in a proportion of ring chromosomes: clues to the mechanisms of formation. J Med Genet 45: 147–154, 2008b. Rossi MR , DiMaio MS, Xiang B, et al. Clinical and genomic characterization of distal duplications and deletions of chromosome 4q: study of two cases and review of the literature. Am J Med Genet 149A : 2788–2794, 2009. Rothenmund H, Chudley AE, Dawson AJ. Familial transmission of a small supernumerary marker chromosome 8 identified by FISH: an update. Am J Med Genet 72: 339–342, 1997. Röthlisberger B, Kotzot D. Recurrence risk in de novo structural chromosomal rearrangements. Am J Med Genet 143A : 1708–1714, 2007. Röthlisberger B, Kotzot D, Brecevic L, et al. Recombinant balanced and unbalanced translocations as a consequence of a balanced complex chromosomal rearrangement involving eight breakpoints in four chromosomes. Eur J Hum Genet 7: 873–883, 1999. Rothman BK . Tentative Pregnancy: Prenatal Diagnosis and the Future of Motherhood. London: Pandora Press, 1988.
Rougeulle C, Lalande M. Angelman syndrome: how many genes to remain silent? Neurogenet 1: 229–237, 1998. Rousseau F, Rouillard P, Morel ML, Khandjian EW, Morgan K . Prevalence of carriers of premutationsize alleles of the FMR1 gene—and implications for the population genetics of the fragile X syndrome. Am J Hum Genet 57: 1006–1018, 1995. Roux C, Tripogney C, Morel F, et al. Segregation of chromosomes in sperm of Robertsonian translocation carriers. Cytogenet Genome Res 111: 291–296, 2005. Rovet J, Netley C, Bailey J, Keenan M, Stewart D. Intelligence and achievement in children with extra X aneuploidy: a longitudinal perspective. Am J Med Genet 60: 356–363, 1995. Rowe LR , Lee JY, Rector L, et al. U-type exchange is the most frequent mechanism for inverted duplication with terminal deletion rearrangements. J Med Genet 46: 694–702, 2009. Rowland RE, Edwards LA , Podd JV. Elevated sister chromatid exchange frequencies in New Zealand Vietnam War veterans. Cytogenet Genome Res 116: 248–251, 2007. Rozenberg P, Bussières L, Senat MV. Dépistage de la trisomie 21 en France: le consensus du pire. J Gynecol Obstet Biol Reprod 36: 95–103, 2007. Rubes J, Selevan SG, Evenson DP, et al. Episodic air pollution is associated with increased DNA fragmentation in human sperm without other changes in semen quality. Hum Reprod 20: 2776–2783, 2005. Rubio C, Buendía P, Rodrigo L, et al. Prognostic factors for preimplantation genetic screening in repeated pregnancy loss. Reprod Biomed Online 18: 687–693, 2009. Rubio C, Simón C, Vidal F, et al. Chromosomal abnormalities and embryo development in recurrent miscarriage couples. Hum Reprod 18: 182–188, 2003. Rudnicka AR , Wald NJ, Huttly W, Hackshaw AK . Influence of maternal smoking on the birth prevalence of Down syndrome and on second trimester screening performance. Prenat Diagn 22: 893–897, 2002. Ruggeri A , Dulcetti F, Miozzo M, et al. Prenatal search for UPD 14 and UPD 15 in 83 cases of familial and de novo heterologous Robertsonian translocations. Prenat Diagn 24: 997–1000, 2004. Ruiz-Casares E, Orera M, Henriques-Gil N. Trisomy 21 with a paternal isochromosome 21q, due to a postzygotic misdivision. Ann Génét 44: s56, 2001. Rumpler Y. Meiotic studies in animal models. Ann Génét 44: s18, 2001.
Rushton DI. Examination of products of conception from previable human pregnancies. J Clin Pathol 34: 819–835, 1981. Russell KL, Ming JE, Patel K , Jukofsky L, Magnusson M, Krantz ID. Dominant paternal transmission of Cornelia de Lange syndrome: a new case and review of 25 previously reported familial recurrences. Am J Med Genet 104: 267–276, 2001. Russell LM, Strike P, Browne CE, Jacobs PA . X chromosome loss and ageing. Cytogenet Genome Res 116: 181–185, 2007. Russo S, Finelli P, Recalcati MP, et al. Molecular and genomic characterisation of cryptic chromosomal alterations leading to paternal duplication of the 11p15.5 Beckwith-Wiedemann region. J Med Genet 43: e39, 2006. Rutgers JL, Scully RE. The androgen insensitivity syndrome (testicular feminization): a clinicopathologic study of 43 cases. Int J Gynecol Pathol 10: 126–144, 1991. Ryan MC, Shooter EM, Notterpek L. Aggresome formation in neuropathy models based on peripheral myelin protein 22 mutations. Neurobiol Dis 10: 109–118, 2002. Rybak EA , Bevilacqua K , Veit CR , Klugman SD, Santoro N. Sibling and self ovum donation for sisters with an intermediate FMR1 mutation: what’s a program to do? Fertil Steril 92: 394 e9–12, 2009. Ryynänen M, Heinonen S, Makkonen M, Kajanoja E, Mannermaa A , Pertti K . Feasibility and acceptance of screening for fragile X mutations in low-risk pregnancies. Eur J Hum Genet 7: 212–216, 1999. Saadallah N, Hultén M. A complex three breakpoint translocation involving chromosomes 2, 4, and 9 identified by meiotic investigations of a human male ascertained for subfertility. Hum Genet 71: 312–320, 1985. Saccone S, Federico C, Solovei I, Croquette MF, Della Valle G, Bernardi G. Identification of the gene-richest bands in human prometaphase chromosomes. Chromosome Res 7: 379–386, 1999. Sachs ES, Jahoda MGJ, Los FJ, Pijpers L, Wladimiroff JW. Trisomy 21 mosaicism in gonads with unexpectedly high recurrence risks. Am J Med Genet Suppl 7: 186–188, 1990. Saffery R , Irvine DV, Griffiths B, Kalitsis P, Wordeman L, Choo KHA . Human centromeres and neocentromeres show identical distribution patterns of >20 functionally important kinetochore-associated proteins. Hum Mol Genet 9: 175–185, 2000. Sago H, Chen E, Conte WJ, et al. True trisomy 2 mosaicism in amniocytes and newborn liver
REFERENCES
•
591
associated with multiple system abnormalities. Am J Med Genet 72: 343–346, 1997. Sahoo T, Bacino CA , German JR , et al. Identification of novel deletions of 15q11q13 in Angelman syndrome by array-CGH: molecular characterization and genotype-phenotype correlations. Eur J Hum Genet 15: 943–949, 2007. Sahoo T, Cheung SW, Ward P, et al. Prenatal diagnosis of chromosomal abnormalities using array-based comparative genomic hybridization. Genet Med 8: 719–727, 2006. Sahoo T, del Gaudio D, German JR , et al. Prader-Willi phenotype caused by paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat Genet 40: 719–721, 2008. Saitoh S, Buiting K , Cassidy SB, et al. Clinical spectrum and molecular diagnosis of Angelman and Prader-Willi syndrome patients with an imprinting mutation. Am J Med Genet 68: 195–206, 1997. Saito-Ohara F, Fukuda Y, Ito M, et al. The Xq22 inversion breakpoint interrupted a novel Ras-like GTPase gene in a patient with Duchenne muscular dystrophy and profound mental retardation. Am J Hum Genet 71: 637–645, 2002. Saitta SC, Harris SE, McDonald-McGinn DM, et al. Independent de novo 22q11.2 deletions in first cousins with DiGeorge/velocardiofacial syndrome. Am J Med Genet 124A : 313–317, 2004. Salafsky IS, MacGregor SN, Claussen U, von Eggeling F. Maternal UPD 20 in an infant from a pregnancy with mosaic trisomy 20. Prenat Diagn 21: 860–863, 2001. Salo P, Ignatius J, Simola KOJ, Tahvanainen E, Kaariainen H. Clinical features of nine males with molecularly defined deletions of the Y chromosome long arm. J Med Genet 32: 711–715, 1995. Salomon LJ, Bernard JP, Nizard J, Ville Y. Firsttrimester screening for fetal triploidy at 11 to 14 weeks: a role for fetal biometry. Prenat Diagn 25: 479–483, 2005. Sampson A , de Crespigny LC. Vanishing twins: the frequency of spontaneous fetal reduction of a twin pregnancy. Ultrasound Obstet Gynecol 2: 107–109, 1992. Sánchez JM, Franzi L, Collia F, De Díaz SL, Panal M, Dubner M. Cytogenetic study of spontaneous abortions by transabdominal villus sampling and direct analysis of villi. Prenat Diagn 19: 601–603, 1999. Sandalinas M, Marquez C, Munné S. Spectral karyotyping of fresh, non-inseminated oocytes. Mol Hum Reprod 8: 580–585, 2002.
592
•
REFERENCES
Sandrin-Garcia P, Macedo C, Martelli LR , et al. Recurrent 22q11.2 deletion in a sibship suggestive of parental germline mosaicism in velocardiofacial syndrome. Clin Genet 61: 380–383, 2002. Sanford Hanna JA , Ball S, Pagon RA , Donlan M. Mother to son transmission of del(1) (q42.1q42.3). Am J Med Genet 98: 103–106, 2001. Sanlaville D, Prieur M, de Blois MC, et al. Functional disomy of the Xq28 chromosome region. Eur J Hum Genet 13: 579–585, 2005. Sanlaville D, Schluth-Bolard C, Turleau C. Distal Xq duplication and functional Xq disomy. Orphanet J Rare Dis 4: 4, 2009. Sarno AP, Moorman AJ, Kalousek DK . Partial molar pregnancy with fetal survival: an unusual example of confined placental mosaicism. Obstet Gynecol 82: 716–719, 1993. Sarri C, Gyftodimou J, Avramopoulos D, et al. Partial trisomy 17q22-qter and partial monosomy Xq27-qter in a girl with a de novo unbalanced translocation due to a postzygotic error: case report and review of the literature on partial trisomy 17qter. Am J Med Genet 70: 87–94, 1997. Sasaki T, Tonoki H, Soejima H, Niikawa N. A 4 Mb cryptic deletion associated with inv(8) (q13.1q24.11) in a patient with trichorhinophalangeal syndrome type I. J Med Genet 34: 335–339, 1997. Sato K , Iwakoshi M, Shimokawa O, et al. Angelman syndrome caused by an identical familial 1,487-kb deletion. Am J Med Genet 143: 98–101, 2007. Saura R , Traore W, Taine L, et al. Prenatal diagnosis of trisomy 9. Six cases and a review of the literature. Prenat Diagn 15: 609–614, 1995. Savage AR , Petersen MB, Pettay D, et al. Elucidating the mechanisms of paternal non-disjunction of chromosome 21 in humans. Hum Mol Genet 7: 1221–1227, 1998. Savarirayan R , Bankier A . Acampomelic campomelic dysplasia with de novo 5q;17q reciprocal translocation and severe phenotype. J Med Genet 35: 597–599, 1998. Savva GM, Morris JK , Mutton DE, Alberman E. Maternal age-specific fetal loss rates in Down syndrome pregnancies. Prenat Diagn 26: 499–504, 2006. Savva GM, Walker K , Morris JK . The maternal age-specific live birth prevalence of trisomies 13 and 18 compared to trisomy 21 (Down syndrome). Prenat Diagn 30: 57–64, 2010. Sawyer JR , Swanson CM, Wheeler G, Cunniff C. Chromosome instability in ICF syndrome: formation of micronuclei from multibranched chromosomes 1 demonstrated by fluorescence in
situ hybridization. Am J Med Genet 56: 203–209, 1995. Scambler PJ. 22q11 deletion syndrome: a role for TBX1 in pharyngeal and cardiovascular development. Pediatr Cardiol 31: 378–390, 2010. Schaaf CP, Scott DA , Wiszniewska J, Beaudet AL. Identification of incestuous parental relationships by SNP-based DNA microarrays. Lancet 377: 555–556, 2011. Schaap AHP, van der Pol HG, Boer K , Leschot NJ, Wolf H. Long-term follow-up of infants after transcervical chorionic villus sampling and after amniocentesis to compare congenital abnormalities and health status. Prenat Diagn 22: 598–604, 2002. Schad CR , Kuffel DG, Wyatt WA , et al. Application of fluorescent in situ hybridization with X and Y chromosome specific probes to buccal smear analysis. Am J Med Genet 66: 187–192, 1996. Schecter A , Constable JD. Commentary: Agent Orange and birth defects in Vietnam. Int J Epidemiol 35: 1230–1232, 2006. Scheffer I, Brett EM, Wilson J, Baraitser M. Angelman’s syndrome. J Med Genet 27: 275–277, 1990. Scheuerle A , Heller K , Elder F. Complete trisomy 1q with mosaic Y;1 translocation: a recurrent aneuploidy presenting diagnostic dilemmas. Am J Med Genet 138A : 166–170, 2005. Schiff JD, Palermo GD, Veeck LL, Goldstein M, Rosenwaks Z, Schlegel PN. Success of testicular sperm extraction and intracytoplasmic sperm injection in men with Klinefelter syndrome. J Clin Endocrinol Metab 90: 6263–6267, 2005. Schinzel A . Catalogue of Unbalanced Chromosome Aberrations in Man. Berlin & New York: Walter de Gruyter, 2001. Schinzel AA , Adelsberger PA , Binkert F, Basaran S, Antonarakis SE. No evidence for a paternal interchromosomal effect from analysis of the origin of nondisjunction in Down syndrome patients with concomitant familial chromosome rearrangements. Am J Hum Genet 50: 288–293, 1992. Schinzel AA , Robinson WP, Binkert F, Torresani T, Werder EA . Exclusively paternal X chromosomes in a girl with short stature. Hum Genet 92: 175–178, 1993. Schlaubitz S, Yatsenko SA , Smith LD, et al. Ovotestes and XY sex reversal in a female with an interstitial 9q33.3-q34.1 deletion encompassing NR5A1 and LMX1B causing features of genitopatellar syndrome. Am J Med Genet 143A : 1071–1081, 2007.
Schlessel JS, Brown WT, Lysikiewicz A , Schiff R , Zaslav AL. Monozygotic twins with trisomy 18: a report of discordant phenotype. J Med Genet 27: 640–642, 1990. Schlessinger D, Mandel JL, Monaco AP, Nelson DL, Willard HF. Report and abstracts of the fourth international workshop on human X chromosome mapping 1993. Cytogenet Cell Genet 64: 147–194, 1993. Schluth-Bolard C, Delobel B, Sanlaville D, et al. Cryptic genomic imbalances in de novo and inherited apparently balanced chromosomal rearrangements: array CGH study of 47 unrelated cases. Eur J Med Genet 52: 291–296, 2009. Schluth-Bolard C, Sanlaville D, Labalme A , et al. 17p13.1 microdeletion involving the TP53 gene in a boy presenting with mental retardation but no tumor. Am J Med Genet 152A : 1278–1282, 2010. Schluth-Bolard C, Till M, Rafat A , et al. Monosomy 19pter and trisomy 19q13-qter in two siblings arising from a maternal pericentric inversion: clinical data and molecular characterization. Eur J Med Genet 51: 622–630, 2008. Schmiady H, Neitzel H. Arrest of human oocytes during meiosis I in two sisters of consanguineous parents: first evidence for an autosomal recessive trait in human infertility: case report. Hum Reprod 17: 2556–2559, 2002. Schmickel RD. Contiguous gene syndromes: a component of recognizable syndromes. J Pediatr 109: 231–241, 1986. Schmidt H, Kern W, Giese R , Hallschmid M, Enders A . Intranasal insulin to improve developmental delay in children with 22q13 deletion syndrome: an exploratory clinical trial. J Med Genet 46: 217–222, 2009. Schmidt M, Du Sart D. Functional disomies of the X chromosome influence the cell selection and hence the X inactivation pattern in females with balanced X-autosome translocations: a review of 122 cases. Am J Med Genet 42: 161–169, 1992. Schmitt-Ney M, Thiele H, Kaltwasser P, Bardoni B, Cisternino M, Scherer G. Two novel SRY missense mutations reducing DNA binding identified in XY females and their mosaic fathers. Am J Hum Genet 56: 862–869, 1995. Schmitz D, Henn W, Netzer C. Commentary: no risk, no objections? Ethical pitfalls of cell-free fetal DNA and RNA testing. Br Med J 339: b2690, 2009. Schmutz SM, Pinno E. Morphology alone does not make an isochromosome. Hum Genet 72: 253–255, 1986. Schneider M, Forrester S, Crain V, Kimonis V. A further case of coincidental Prader-Willi and
REFERENCES
•
593
Klinefelter syndromes. Am J Med Genet 126A : 213–214, 2004. Schon EA , Kim SH, Ferreira JC, et al. Chromosomal non-disjunction in human oocytes: is there a mitochondrial connection? Hum Reprod 15(suppl 2): 160–172, 2000. Schönherr N, Meyer E, Roos A , Schmidt A , Wollmann HA , Eggermann T. The centromeric 11p15 imprinting centre is also involved in Silver-Russell syndrome. J Med Genet 44: 59–63, 2007. Schoolcraft WB, Keller JL, Schlenker T. Excellent embryo quality obtained from vitrified oocytes. Reprod Biomed Online 19: 820–823, 2009. Schorderet DF, Friedman C, Disteche CM. Pericentric inversion of the X chromosome: presentation of a case and review of the literature. Ann Génét 34: 98–103, 1991. Schouten JP, McElgunn CJ, Waaijer R , Zwijnenburg D, Diepvens F, Pals G. Relative quantification of 40 nucleic acid sequences by multiplex ligationdependent probe amplification. Nucleic Acids Res 30: e57, 2002. Schull WJ, Neel JV. Maternal radiation and mongolism. Lancet 279: 537–538, 1962. Schultz BAH, Roberts S, Rodgers A, Ataya K . Pregnancy in true hermaphrodites and all male offspring to date. Obstet Gynecol 113: 534–536, 2009. Schulz S, Gerloff C, Ledig S, et al. Prenatal diagnosis of Roberts syndrome and detection of an ESCO2 frameshift mutation in a Pakistani family. Prenat Diagn 28: 42–45, 2008. Schuring-Blom GH, Boer K , Knegt AC, Verjaal M, Leschot NJ. Trisomy 13 or 18 (mosaicism) in first trimester cytotrophoblast cells: false-positive results in 11 out of 51 cases. Eur J Obstet Gynecol Reprod Biol 101: 161–168, 2002. Schuurs-Hoeijmakers JHM, Vermeer S, van Bon BWM, et al. Refining the critical region of the novel 19q13.11 microdeletion syndrome to 750 Kb. J Med Genet 46: 421–423, 2009. Schwartz S, Depinet TW, Leana-Cox J, et al. Sex chromosome markers: characterization using fluorescence in situ hybridization and review of the literature. Am J Med Genet 71: 1–7, 1997. Schwartz S, Raffel LJ. Prenatal detection of 45,X/46,XX/47,XXX mosaicism through amniocentesis: mosaicism confirmed in cord blood, amnion, and chorion. Prenat Diagn 12: 1043–1046, 1992. Schwarzbraun T, Obenauf AC, Langmann A , et al. Predictive diagnosis of the cancer prone Li-Fraumeni syndrome by accident: new challenges through whole genome array testing. J Med Genet 46: 341–344, 2009.
594
•
REFERENCES
Schwendemann WD, Contag SA , Wax JR , et al. Sonographic findings in trisomy 9. J Ultrasound Med 28: 39–42, 2009. Schwinger E, Mikkelsen M, Niesen M. Familial balanced (7;11;21) translocation and Down’s syndrome in two siblings. Clin Genet 7: 304–307, 1975. Scott JA , Wenger SL, Steele MW, Chakravarti A . Down syndrome consequent to a cryptic maternal 12p;21q chromosome translocation. Am J Med Genet 56: 67–71, 1995. Scott RH, Douglas J, Baskcomb L, et al. Constitutional 11p15 abnormalities, including heritable imprinting center mutations, cause nonsyndromic Wilms tumor. Nat Genet 40: 1329–1334, 2008a. Scott RH, Douglas J, Baskcomb L, et al. Methylationspecific MLPA (MS-MLPA) robustly detects and distinguishes 11p15 abnormalities associated with overgrowth and growth retardation. J Med Genet 45: 106–113, 2008b. Scriven PN, Handyside AH, Mackie Ogilvie C. Chromosome translocations: segregation modes and strategies for preimplantation genetic diagnosis. Prenat Diagn 18: 1437–1449, 1998. Sebat J, Lakshmi B, Troge J, et al. Large-scale copy number polymorphism in the human genome. Science 305: 525–528, 2004. Sebire NJ, Lindsay I. p57KIP2 immunostaining in the diagnosis of complete versus partial hydatidiform moles. Histopathol 48: 873–874, 2006. Sebire NJ, Snijders RJ, Santiago C, Papapanagiotou G, Nicolaides KH. Management of twin pregnancies with fetal trisomies. Br J Obstet Gynaecol 104: 220–222, 1997. Seckl MJ, Fisher RA , Salerno G, et al. Choriocarcinoma and partial hydatidiform moles. Lancet 356: 36–39, 2000. Seeherunvong T, Perera EM, Bao Y, et al. 46,XX sex reversal with partial duplication of chromosome arm 22q. Am J Med Genet 127A : 149–151, 2004. Seemanová E, Sperling K , Neitzel H, et al. Nijmegen breakage syndrome (NBS) with neurological abnormalities and without chromosomal instability. J Med Genet 43: 218–224, 2006. Seghezzi L, Maserati E, Minelli A , et al. Constitutional trisomy 8 as first mutation in multistep carcinogenesis: clinical, cytogenetic, and molecular data on three cases. Genes Chromosomes Cancer 17: 94–101, 1996. Seidel J, Schiller S, Kelbova C, et al. Brachytelephalangic dwarfism due to the loss of ARSE and SHOX genes resulting from an X;Y translocation. Clin Genet 59: 115–121, 2001.
Seifer DB, DeJesus V, Hubbard K . Mitochondrial deletions in luteinized granulosa cells as a function of age in women undergoing in vitro fertilization. Fertil Steril 78: 1046–1048, 2002. Selicorni A , Russo S, Gervasini C, et al. Clinical score of 62 Italian patients with Cornelia de Lange syndrome and correlations with the presence and type of NIPBL mutation. Clin Genet 72: 98–108, 2007. Seller MJ, Mazzaschi R , Ogilvie CM, Mohammed S. A trisomy 2 fetus with severe neural tube defects and other abnormalities. Clin Dysmorphol 13: 25–27, 2004. Selvaraj K , Ganesh V, Selvaraj P. Successful pregnancy in a patient with a 46,XY karyotype. Fertil Steril 78: 419–420, 2002. Sepulveda W, Be C, Youlton R , Carstens E, Reyes M. Nuchal translucency thickness and outcome in chromosome translocation diagnosed in the first trimester. Prenat Diagn 21: 726–728, 2001. Sepulveda W, Wong AE, Dezerega V. First-trimester sonographic findings in trisomy 18: a review of 53 cases. Prenat Diagn 30: 256–259, 2010. Serra A , Bova R . Acrocentric chromosome double NOR is not a risk factor for Down syndrome. Am J Med Genet Suppl 7: 169–174, 1990. Seven M, Karaman B, Hacihanefioglu S, Deviren A , Yuksel A , Basaran S. Neuroblastoma in a dysmorphic girl with a partial duplication of 2p caused by an unbalanced translocation. Clin Dysmorphol 11: 39–42, 2002. Sévin M, Kutalik Z, Bergman S, et al. Penetrance of marked cognitive impairment in older male carriers of the FMR1 gene premutation. J Med Genet 46: 818–824, 2009. Shaffer BL, Caughey AB, Norton ME. Variation in the decision to terminate pregnancy in the setting of fetal aneuploidy. Prenat Diagn 26: 667–671, 2006a. Shaffer LG. American College of Medical Genetics guideline on the cytogenetic evaluation of the individual with developmental delay or mental retardation. Genet Med 7: 650–654, 2005. Shaffer LG. Risk estimates for uniparental disomy following prenatal detection of a nonhomologous Robertsonian translocation. Prenat Diagn 26: 303–307, 2006. Shaffer LG, Agan N, Goldberg JD, Ledbetter DH, Longshore JW, Cassidy SB. American College of Medical Genetics statement of diagnostic testing for uniparental disomy. Genet Med 3: 206–211, 2001. Shaffer LG, Bejjani BA . Medical applications of array CGH and the transformation of clinical cytogenetics. Cytogenet Genome Res 115: 303–309, 2006.
Shaffer LG, Bejjani BA , Torchia B, Kirkpatrick S, Coppinger J, Ballif BC. The identification of microdeletion syndromes and other chromosome abnormalities: cytogenetic methods of the past, new technologies for the future. Am J Med Genet 145C: 335–345, 2007a. Shaffer LG, Bui TH. Molecular cytogenetic and rapid aneuploidy detection methods in prenatal diagnosis. Am J Med Genet 145C: 87–98, 2007. Shaffer LG, Coppinger J, Alliman S, et al. Comparison of microarray-based detection rates for cytogenetic abnormalities in prenatal and neonatal specimens. Prenat Diagn 28: 789–795, 2008. Shaffer LG, Hecht JT, Ledbetter DH, Greenberg F. Familial interstitial deletion 11(p11.12p12) associated with parietal foramina, brachymicrocephaly, and mental retardation. Am J Med Genet 45: 581–583, 1993. Shaffer LG, Jackson-Cook CK , Meyer JM, Brown JA , Spence JE. A molecular genetic approach to the identification of isochromosomes of chromosome 21. Hum Genet 86: 375–382, 1991. Shaffer LG, Kashork CD, Saleki R , et al. Targeted genomic microarray analysis for identification of chromosome abnormalities in 1500 consecutive clinical cases. J Pediatr 149: 98–102, 2006b. Shaffer LG, Kennedy GM, Spikes AS, Lupski JR . Diagnosis of CMT1A duplications and HNPP deletions by interphase FISH: implications for testing in the cytogenetics laboratory. Am J Med Genet 69: 325–331, 1997a. Shaffer LG, Lupski JR . Molecular mechanisms for constitutional chromosomal rearrangements in humans. Annu Rev Genet 34: 297–329, 2000. Shaffer LG, McCaskill C, Adkins K , Hassold TJ. Systematic search for uniparental disomy in early fetal losses: the results and a review of the literature. Am J Med Genet 79: 366–372, 1998. Shaffer LG, McCaskill C, Egli CA , Baker JC, Johnston KM. Is there an abnormal phenotype associated with maternal isodisomy for chromosome 2 in the presence of two isochromosomes? Am J Hum Genet 61: 461–462, 1997b. Shaffer LG, Theisen A , Bejjani BA , et al. The discovery of microdeletion syndromes in the post-genomic era: review of the methodology and characterization of a new 1q41q42 microdeletion syndrome. Genet Med 9: 607–616, 2007b. Shaikh TH, Gai X , Perin JC, et al. High-resolution mapping and analysis of copy number variations in the human genome: a data resource for clinical and research applications. Genome Res 19: 1682–1690, 2009. Shalev E, Zalel Y, Weiner E, Cohen H, Shneur Y. The role of cordocentesis in assessment of mosaicism
REFERENCES
•
595
found in amniotic fluid cell culture. Acta Obstet Gynecol Scand 73: 119–122, 1994. Shanske A , Ellison J, Vuguin P, et al. Deletion of the pseudoautosomal region in a male with a unique Y;13 translocation and short stature. Am J Med Genet 82: 34–39, 1999a. Shanske A , Ferreira JC, Leonard JC, Fuller P, Marion RW. Hirschsprung disease in an infant with a contiguous gene syndrome of chromosome 13. Am J Med Genet 102: 231–236, 2001. Shanske AL, Dowling P, Schmidt R , et al. Simultaneous occurrence of two supernumerary autosomal ring chromosomes r(1) and r(16) in twins. J Med Genet 36: 625–628, 1999b. Shao Y, Cuccaro ML, Hauser ER , et al. Fine mapping of autistic disorder to chromosome 15q11-q13 by use of phenotypic subtypes. Am J Hum Genet 72: 539–548, 2003. Shapiro BL. Whither Down syndrome critical regions? Hum Genet 99: 421–423, 1997. Shapira M, Dar H, Bar-El H, Bar-Nitzan N, Even L, Borochowitz Z. Inherited inverted duplication of X chromosome in a male: report of a patient and review of the literature. Am J Med Genet 72: 409–414, 1997. Shapira SK , McCaskill C, Northrup H, et al. Chromosome 1p36 deletions: the clinical phenotype and molecular characterization of a common newly delineated syndrome. Am J Hum Genet 61: 642–650, 1997. Shapira SK , Orr-Urtreger A , Gagos S, Shaffer LG. Constitutional mosaicism for a chromosome 9 inversion resulting in recombinant aneusomy in an offspring. Am J Med Genet 69: 360–364, 1997c. Sharp A , Kusz K , Jaruzelska J, Szarras-Czapnik M, Wolski J, Jacobs P. Familial X/Y translocations associated with variable sexual phenotype. J Med Genet 41: 440–444, 2004. Sharp A , Robinson D, Jacobs P. Age- and tissuespecific variation of X chromosome inactivation ratios in normal women. Hum Genet 107: 343–349, 2000. Sharp AJ. Emerging themes and new challenges in defining the role of structural variation in human disease. Hum Mutat 30: 135–144, 2009. Sharp AJ, Mefford HC, Li K , et al. A recurrent 15q13.3 microdeletion syndrome associated with mental retardation and seizures. Nat Genet 40: 322–328, 2008. Shashi V, Golden WL, Allinson PS, Blanton SH, von Kap-Herr C, Kelly TE. Molecular analysis of recombination in a family with Duchenne muscular dystrophy and a large pericentric X chromosome inversion. Am J Hum Genet 58: 1231–1238, 1996.
596
•
REFERENCES
Shaw MA , Chiurazzi P, Romain DR , Neri G, Gécz J. A novel gene, FAM11A, associated with the FRAXF CpG island is transcriptionally silent in FRAXF full mutation. Eur J Hum Genet 10: 767–772, 2002. Sheath KL, Asquith PM, Zhang L, Aftimos S. Prenatal diagnosis of trisomy 3 mosaicism in a fetus with severe IUGR . Prenat Diagn 30: 803–805, 2010. Sherard J, Bean C, Bove B, et al. Long survival in a 69,XXY triploid male. Am J Med Genet 25: 307–312, 1986. Sheridan E, Williams J, Caine A , Morgan R , Mason G, Mueller RF. Counselling implications of chromosomal abnormalities other than trisomy 21 detected through a maternal serum screening programme. Br J Obstet Gynaecol 104: 42–45, 1997. Sheridan MB, Kato T, Haldeman-Englert C, et al. A palindrome-mediated recurrent translocation with 3:1 meiotic nondisjunction: the t(8;22) (q24.13;q11.21). Am J Hum Genet 13; 87: 209–218, 2010. Sherman SL, Iselius L, Gallano P, et al. Segregation analysis of balanced pericentric inversions in pedigree data. Clin Genet 30: 87–94, 1986. Sherman SL, Petersen MB, Freeman SB, et al. Non-disjunction of chromosome 21 in maternal meiosis I: evidence for a maternal age-dependent mechanism involving reduced recombination. Hum Mol Genet 3: 1529–1535, 1994. Sheth J, Joshi R , Sheth F. Ring chromosome 9 in a dysmorphic child. Indian J Pediatr 74: 507–508, 2007. Shevell M, Ashwal S, Donley D, et al. Practice parameter: evaluation of the child with global developmental delay: report of the quality standards subcommittee of the American Academy of Neurology and the practice committee of the Child Neurology Society. Neurology 60: 367–380, 2003. Shi Q, Ko E, Barclay L, Hoang T, Rademaker A , Martin R . Cigarette smoking and aneuploidy in human sperm. Mol Reprod Dev 59: 417–421, 2001a. Shi Q, Martin RH. Spontaneous frequencies of aneuploid and diploid sperm in 10 normal Chinese men: assessed by multicolor fluorescence in situ hybridization. Cytogenet Cell Genet 90: 79–83, 2000a. Shi Q, Martin RH. Aneuploidy in human sperm: a review of the frequency and distribution of aneuploidy, effects of donor age and lifestyle factors. Cytogenet Cell Genet 90: 219–226, 2000b. Shi Q, Martin RH. Aneuploidy in human spermatozoa: FISH analysis in men with
constitutional chromosomal abnormalities, and in infertile men. Reproduction 121: 655–666, 2001. Shi Q, Schmid TE, Adler I. Griseofulvin-induced aneuploidy and meiotic delay in male mouse germ cells: detected by using conventional cytogenetics and three-color FISH. Mutat Res 441: 181–190, 1999. Shi Q, Spriggs E, Field LL, Ko E, Barclay L, Martin RH. Single sperm typing demonstrates that reduced recombination is associated with the production of aneuploid 24,XY human sperm. Am J Med Genet 99: 34–38, 2001b. Shi Q, Spriggs E, Field LL, et al. Absence of age effect on meiotic recombination between human X and Y chromosomes. Am J Hum Genet 71: 254–261, 2002. Shim SH, Lee CH, Lee JY, et al. A de novo centric fission of chromosome 11 in a patient with recurrent miscarriages. J Korean Med Sci 22: 146–148, 2007. Shimokawa O, Kurosawa K , Ida T, et al. Molecular characterization of inv dup del(8p): analysis of five cases. Am J Med Genet 128A : 133–137, 2004. Shinawi M, Liu P, Kang SH, et al. Recurrent reciprocal 16p11.2 rearrangements associated with global developmental delay, behavioural problems, dysmorphism, epilepsy, and abnormal head size. J Med Genet 47: 332–341, 2010. Shobha Rani A , Jyothi A , Reddy PP, Reddy OS. Reproduction in Down’s syndrome. Int J Gynaecol Obstet 31: 81–86, 1990. Shohat M, Legum C, Romem Y, Borochowitz Z, Bach G, Goldman B. Down syndrome prevention program in a population with an older maternal age. Obstet Gynecol 85: 368–373, 1995. Shuib S, McMullan D, Rattenberry E, et al. Microarray based analysis of 3p25-p26 deletions (3psyndrome). Am J Med Genet 149A : 2099–2105, 2009. Shuster E. Microarray genetic screening: a prenatal roadblock for life? Lancet 369: 526–529, 2007. Shuttleworth GE. Mongolian imbecility. Br Med J 2: 661–665, 1909. Sifakis S, Ghatpande S, Seppo A , et al. Prenatal diagnosis of trisomy 21 through detection of trophoblasts in cervical smears. Early Hum Dev 86: 311–313, 2010. Sifakis S, Karkaletsi M, Christopoulou S, et al. Distinctive pattern of first trimester maternal serum biochemical markers in trisomy 22 pregnancies. Prenat Diagn 28: 1174–1176, 2008. Siggberg L, Ala-Mello S, Jaakkola E, et al. Array CGH in molecular diagnosis of mental retardation - A study of 150 Finnish patients. Am J Med Genet 152A : 1398–1410, 2010.
Sigurdardottir S, Goodman BK , Rutberg J, Thomas GH, Jabs EW, Geraghty MT. Clinical, cytogenetic, and fluorescence in situ hybridization findings in two cases of “complete ring” syndrome. Am J Med Genet 87: 384–390, 1999. Sigurdson AJ, Bhatti P, Preston DL, et al. Routine diagnostic X-ray examinations and increased frequency of chromosome translocations among U.S. radiologic technologists. Cancer Res 68: 8825–8831, 2008. Sijmons RH, Leegte B, van Lingen RA, et al. Tetrasomy 5p mosaicism in a boy with delayed growth, hypotonia, minor anomalies, and an additional isochromosome 5p [46,XY/47,XY, + i(5p)]. Am J Med Genet 47: 559–562, 1993. Sikkema-Raddatz B, Bouman K , VerschuurenBemelmans CC, de Jong B. Trisomy 12 mosaicism in CVS culture confirmed in the fetus. Prenat Diagn 19: 1176–1177, 1999. Sikkema-Raddatz B, Castedo S, Te Meerman GJ. Probability tables for exclusion of mosaicism in prenatal diagnosis. Prenat Diagn 17: 115–118, 1997a. Sikkema-Raddatz B, Verschuuren-Bemelmans CC, Kloosterman M, de Jong B. A 46,XX,der(13;14) (q10;q10),+21 child born after a 45,XX,der(13;14)(q10;q10) chromosomal finding in CVS. Prenat Diagn 17: 1086–1088, 1997b. Silber S, Escudero T, Lenahan K , Abdelhadi I, Kilani Z, Munné S. Chromosomal abnormalities in embryos derived from testicular sperm extraction. Fertil Steril 79: 30–38, 2003. Silverstein S, Lerer I, Sagi M, Frumkin A , Ben-Neriah Z, Abeliovich D. Uniparental disomy in fetuses diagnosed with balanced Robertsonian translocations: risk estimate. Prenat Diagn 22: 649–651, 2002. Simmers RN, Sutherland GR , West A , Richards RI. Fragile sites at 16q22 are not at the breakpoint of the chromosomal rearrangement in AMMoL. Science 236: 92–94, 1987. Simpson JL, Meyers CM, Martin AO, Elias S, Ober C. Translocations are infrequent among couples having repeated spontaneous abortions but no other abnormal pregnancies. Fertil Steril 51: 811–814, 1989. Simpson JL, Rajkovic A . Ovarian differentiation and gonadal failure. Am J Med Genet 89: 186–200, 1999. Singh R , Gardner RJM, Crossland KM, Scheffer IE, Berkovic SF. Chromosomal abnormalities and epilepsy: a review for clinicians and gene hunters. Epilepsia 43: 127–140, 2002.
REFERENCES
•
597
Singh SM, Murphy B, O’Reilly R . Monozygotic twins with chromosome 22q11 deletion and discordant phenotypes: updates with an epigenetic hypothesis. J Med Genet 39: e71, 2002. Sirchia SM, Garagiola I, Colucci G, et al. Trisomic zygote rescue revealed by DNA polymorphism analysis in confined placental mosaicism. Prenat Diagn 18: 201–206, 1998. Sismani C, Kitsiou-Tzeli S, Ioannides M, et al. Cryptic genomic imbalances in patients with de novo or familial apparently balanced translocations and abnormal phenotype. Mol Cytogenet 1: 15, 2008. Sivak LE, Esbenshade J, Brothman AR , Issa B, Lemons RS, Carey JC. Multiple congenital anomalies in a man with (X;6) translocation. Am J Med Genet 51: 9–12, 1994. Sivertsen A , Lie RT, Wilcox AJ, et al. Prevalence of duplications and deletions of the 22q11 DiGeorge syndrome region in a population-based sample of infants with cleft palate. Am J Med Genet 143: 129–134, 2007. Skakkebæk NE, Giwercman A , de Kretser D. Pathogenesis and management of male infertility. Lancet 343: 1473–1479, 1994. Sklower Brooks SS, Genovese M, Gu H, Duncan CJ, Shanske A , Jenkins EC. Normal adaptive function with learning disability in duplication 8p including band p22. Am J Med Genet 78: 114–117, 1998. Skotko BG, Kishnani PS, Capone GT. Prenatal diagnosis of Down syndrome: how best to deliver the news. Am J Med Genet 149A : 2361–2367, 2009. Skuse DH, James RS, Bishop DVM, et al. Evidence from Turner’s syndrome of an imprinted X-linked locus affecting cognitive function. Nature 387: 705–708, 1997. Slaney SF, Chalmers IJ, Affara NA , Chitty LS. An autosomal or X linked mutation results in true hermaphrodites and 46,XX males in the same family. J Med Genet 35: 17–22, 1998. Slater H, Shaw JH, Bankier A , Forrest SM, Dawson G. UPD 13: no indication of maternal or paternal imprinting of genes on chromosome 13. J Med Genet 32: 493, 1995. Slater H, Shaw JH, Dawson G, Bankier A , Forrest SM. Maternal uniparental disomy of chromosome 13 in a phenotypically normal child. J Med Genet 31: 644–646, 1994. Slater HR , Nouri S, Earle E, Lo AW, Hale LG, Choo KHA . Neocentromere formation in a stable ring 1p32-p36.1 chromosome. J Med Genet 36: 914–918, 1999. Slater HR , Ralph A , Daniel A , Worthington S, Roberts C. A case of maternal uniparental disomy of chromosome 9 diagnosed prenatally and the
598
•
REFERENCES
related problem of residual trisomy. Prenat Diagn 20: 930–932, 2000. Slavotinek A , Gaunt L, Donnai D. Paternally inherited duplications of 11p15.5 and Beckwith-Wiedemann syndrome. J Med Genet 34: 819–826, 1997. Slavotinek A , Lee SS, Davis R , et al. Fryns syndrome phenotype caused by chromosome microdeletions at 15q26.2 and 8p23.1. J Med Genet 42: 730–736, 2005. Slavotinek AM. Novel microdeletion syndromes detected by chromosome microarrays. Hum Genet 124: 1–17, 2008. Slim R , Ao A , Surti U, et al. Recurrent triploid and dispermic conceptions in patients with NLRP7 mutations. Placenta 32: 409–412, 2011. Slim R , Mehio A . The genetics of hydatidiform moles: new lights on an ancient disease. Clin Genet 71: 25–34, 2007. Sloter E, Nath J, Eskenazi B, Wyrobek AJ. Effects of male age on the frequencies of germinal and heritable chromosomal abnormalities in humans and rodents. Fertil Steril 81: 925–943, 2004. Smeets DFCM, Hamel BCJ, Nelen MR , et al. Prader-Willi syndrome and Angelman syndrome in cousins from a family with a translocation between chromosomes 6 and 15. N Engl J Med 326: 807–811, 1992. Smith A , Deng ZM, Beran R , Woodage T, Trent RJ. Familial unbalanced translocation t(8;15) (p23.3;q11) with uniparental disomy in Angelman syndrome. Hum Genet 93: 471–473, 1994. Smith A , Robson L. Low frequency of inherited deletions of 22q11. Am J Med Genet 85: 513–514, 1999. Smith A , Watt AJ, Cummins M, Gardner RJM, Wilson M. A small one-band paracentric inversion inv (4) (p15.3p16.3). Ann Génét 35: 161–163, 1992. Smith ACM, Dykens E, Greenberg F. Behavioral phenotype of Smith-Magenis syndrome (del 17p11.2). Am J Med Genet 81: 179–185, 1998a. Smith ACM, Spuhler K , Williams TM, McConnell T, Sujansky E, Robinson A . Genetic risk for recombinant 8 syndrome and the transmission rate of balanced inversion 8 in the Hispanic population of the southwestern United States. Am J Hum Genet 41: 1083–1103, 1987. Smith K , Lowther G, Maher E, Hourihan T, Wilkinson T, Wolstenholme J. The predictive value of findings of the common aneuploidies, trisomies 13, 18 and 21, and numerical sex chromosome abnormalities at CVS: experience from the ACC U.K . Collaborative Study. Association of Clinical Cytogeneticists Prenatal Diagnosis Working Party. Prenat Diagn 19: 817–826, 1999.
Smith MJ, Creasy MR , Clarke A , Upadhyaya M. Sex ratio and absence of uniparental disomy in spontaneous abortions with a normal karyotype. Clin Genet 53: 258–261, 1998b. Smrcek JM, Germer U, Krokowski M, et al. Prenatal ultrasound diagnosis and management of body stalk anomaly: analysis of nine singleton and two multiple pregnancies. Ultrasound Obstet Gynecol 21: 322–328, 2003. Snijders AM, Nowak N, Segraves R , et al. Assembly of microarrays for genome-wide measurement of DNA copy number. Nat Genet 29: 263–264, 2001. Snijders RJM, Sebire NJ, Nicolaides KH. Maternal age and gestational age-specific risk for chromosomal defects. Fetal Diagn Ther 10: 356–367, 1995. Soares SR , Templado C, Blanco J, Egozcue J, Vidal F. Numerical chromosome abnormalities in the spermatozoa of the fathers of children with trisomy 21 of paternal origin: generalised tendency to meiotic non-disjunction. Hum Genet 108: 134–139, 2001a. Soares SR , Vidal F, Bosch M, et al. Acrocentric chromosome disomy is increased in spermatozoa from fathers of Turner syndrome patients. Hum Genet 108: 499–503, 2001b. Sodré CP, Guilherme RS, Meloni VF, et al. Ring chromosome instability evaluation in six patients with autosomal rings. Genet Mol Res 9: 134–143, 2010. Soler A , Margarit E, Queralt R , et al. Paternal isodisomy 13 in a normal newborn infant after trisomy rescue evidenced by prenatal diagnosis. Am J Med Genet 90: 291–293, 2000. Soler A , Salami C, Balmes I, et al. Pericentric X chromosome in a family. Clin Genet 20: 234–235, 1981. Soler A , Sánchez A , Carrió A , et al. Recombination in a male carrier of two reciprocal translocations involving chromosomes 14, 14′, 15, and 21 leading to balanced and unbalanced rearrangements in offspring. Am J Med Genet 134A : 309–314, 2005. Somers CM, Cooper DN. Air pollution and mutations in the germline: are humans at risk? Hum Genet 125: 119–130, 2009. Somprasit C, Aguinaga M, Cisneros PL, et al. Paternal gonadal mosaicism detected in a couple with recurrent abortions undergoing PGD: FISH analysis of sperm nuclei proves valuable. Reprod Biomed Online 9: 225–230, 2004. Sonntag B, Meschede D, Ullmann V, et al. Low-level sex chromosome mosaicism in female partners of couples undergoing ICSI therapy does not significantly affect treatment outcome. Hum Reprod 16: 1648–1652, 2001.
Souka AP, Von Kaisenberg CS, Hyett JA , Sonek JD, Nicolaides KH. Increased nuchal translucency with normal karyotype. Am J Obstet Gynecol 192: 1005–1021, 2005. South ST, Lamb AN. Detecting genomic imbalances in prenatal diagnosis: main hurdles and recent advances. Expert Opin Med Diagn 3: 227–235, 2009. South ST, Rope AF, Lamb AN, et al. Expansion in size of a terminal deletion: a paradigm shift for parental follow-up studies. J Med Genet 45: 391–395, 2008a. South ST, Swensen JJ, Maxwell T, Rope A , Brothman AR , Chen Z. A new genomic mechanism leading to cri-du-chat syndrome. Am J Med Genet 140: 2714–2720, 2006. South ST, Whitby H, Battaglia A , Carey JC, Brothman AR . Comprehensive analysis of Wolf-Hirschhorn syndrome using array CGH indicates a high prevalence of translocations. Eur J Hum Genet 16: 45–52, 2008b. South ST, Whitby H, Maxwell T, Aston E, Brothman AR , Carey JC. Co-occurrence of 4p16.3 deletions with both paternal and maternal duplications of 11p 15: modification of the Wolf-Hirschhorn syndrome phenotype by genetic alterations predicted to result in either a BeckwithWiedemann or Russell-Silver phenotype. Am J Med Genet 146A : 2691–2697, 2008c. Soysal Y, Balci S, Hekimler K , et al. Characterization of double ring chromosome 4 mosaicism associated with bilateral hip dislocation, cortical dysgenesis, and epilepsy. Am J Med Genet 149A : 2782–2787, 2009. Spak DK , Johnston K , Donlon TA . A noncentromeric C band variant on chromosome 11q23.2. J Med Genet 26: 532–534, 1989. Spandorfer SD, Chung PH, Kligman I, Liu HC, Davis OK , Rosenwaks Z. An analysis of the effect of age on implantation rates. J Assist Reprod Genet 17: 303–306, 2000. Sparago A , Cerrato F, Vernucci M, Ferrero GB, Silengo MC, Riccio A . Microdeletions in the human H19 DMR result in loss of IGF2 imprinting and Beckwith-Wiedemann syndrome. Nat Genet 36: 958–960, 2004. Sparkes RL, Shetty S, Chernos JE, Mefford HC, Micheil Innes A . Interstitial deletion of 11q in a mother and fetus: implications of directly transmitted chromosomal imbalances for prenatal genetic counseling. Prenat Diagn 29: 283–286, 2009. Speed RM. Meiotic configurations in female trisomy 21 foetuses. Hum Genet 66: 176–180, 1984.
REFERENCES
•
599
Speed RM. The possible role of meiotic pairing anomalies in the atresia of human fetal oocytes. Hum Genet 78: 260–266, 1988. Speevak M, Farrell SA , Chadwick D. Molecular and cytogenetic characterization of a prenatally ascertained de novo (X;Y) translocation. Am J Med Genet 98: 107–108, 2001. Speevak MD, McGowan-Jordan J, Chun K. The detection of chromosome anomalies by QF-PCR and residual risks as compared to G-banded analysis. Prenat Diagn 31: 454–458, 2011. Speevak MD, Smart C, Unwin L, Bell M, Farrell SA . Molecular characterization of an inherited ring (19) demonstrating ring opening. Am J Med Genet 121A : 141–145, 2003. Spencer K . What is the true fetal loss rate in pregnancies affected by trisomy 21 and how does this influence whether first trimester detection rates are superior to those in the second trimester? Prenat Diagn 21: 788–789, 2001. Spencer K . Aneuploidy screening in the first trimester. Am J Med Genet 145C: 18–32, 2007. Spencer K , Kagan KO, Nicolaides KH. Screening for trisomy 21 in twin pregnancies in the first trimester: an update of the impact of chorionicity on maternal serum markers. Prenat Diagn 28: 49–52, 2008. Spencer K , Staboulidou I, Nicolaides KH. First trimester aneuploidy screening in the presence of a vanishing twin: implications for maternal serum markers. Prenat Diagn 30: 235–240, 2010. Staals JE, Schrander-Stumpel CT, Hamers G, Fryns JP. Prenatal diagnosis of trisomy 12 mosaicism: normal development of a 3 years old female child. Genet Couns 14: 233–237, 2003. Staebler M, Donner C, Van Regemorter N, et al. Should determination of the karyotype be systematic for all malformations detected by obstetrical ultrasound? Prenat Diagn 25: 567–573, 2005. Staessen C, Platteau P, Van Assche E, et al. Comparison of blastocyst transfer with or without preimplantation genetic diagnosis for aneuploidy screening in couples with advanced maternal age: a prospective randomized controlled trial. Hum Reprod 19: 2849–2858, 2004. Staessen C, Tournaye H, Van Assche E, et al. PGD in 47,XXY Klinefelter’s syndrome patients. Hum Reprod Update 9: 319–330, 2003. Stalker HJ, Gray BA , Zori RT. Dominant transmission of a previously unidentified 13/17 translocation in a five-generation family with Robin cleft and other skeletal defects. Am J Med Genet 103: 339–341, 2001.
600
•
REFERENCES
Stalker HJ, Williams CA . Genetic counseling in Angelman syndrome: the challenges of multiple causes. Am J Med Genet 77: 54–59, 1998. Stalker HJ, Williams CA , Wagstaff J. Genetic counseling in Angelman syndrome: gonadal mosaicism. Am J Med Genet 78: 482, 1998. Stallard R , Krueger S, James RS, Schwartz S. Uniparental isodisomy 13 in a normal female due to transmission of a maternal t(13q13q). Am J Med Genet 57: 14–18, 1995. Stamberg J, Thomas GH. Unusual supernumerary chromosomes: types encountered in a referred population, and high incidence of associated maternal chromosome abnormalities. Hum Genet 72: 140–144, 1986. Stankiewicz P, Brozek I, Hélias-Rodzewicz Z, et al. Clinical and molecular-cytogenetic studies in seven patients with ring chromosome 18. Am J Med Genet 101: 226–239, 2001a. Stankiewicz P, Kuechler A , Eller CD, et al. Minimal phenotype in a girl with trisomy 15q due to t(X;15)(q22.3;q11.2) translocation. Am J Med Genet 140: 442–452, 2006. Stankiewicz P, Rujner J, Löffler C, et al. Alagille syndrome associated with a paracentric inversion 20p12.2p13 disrupting the JAG1 gene. Am J Med Genet 103: 166–171, 2001b. Stankiewicz P, Sen P, Bhatt SS, et al. Genomic and genic deletions of the FOX gene cluster on 16q24.1 and inactivating mutations of FOXF1 cause alveolar capillary dysplasia and other malformations. Am J Hum Genet 84: 780–791, 2009. Stankiewicz P, Thiele H, Baldermann C, et al. Phenotypic findings due to trisomy 7p15.3-pter including the TWIST locus. Am J Med Genet 103: 56–62, 2001c. Stankiewicz P, Thiele H, Schlicker M, et al. Duplication of Xq26.2-q27.1, including SOX3, in a mother and daughter with short stature and dyslalia. Am J Med Genet 138: 11–17, 2005. Staples AJ, Sutherland GR , Haan EA , Clisby S. Epidemiology of Down syndrome in South Australia, 1960–89. Am J Hum Genet 49: 1014–1024, 1991. Stark KL, Xu B, Bagchi A , et al. Altered brain microRNA biogenesis contributes to phenotypic deficits in a 22q11-deletion mouse model. Nat Genet 40: 751–760, 2008. Starke H, Seidel J, Henn W, et al. Homologous sequences at human chromosome 9 bands p12 and q13–21.1 are involved in different patterns of pericentric rearrangements. Eur J Hum Genet 10: 790–800, 2002.
Stasiewicz-Jarocka B, Raczkiewicz B, Kowalczyk D, Zawada M, Midro AT. Genetic risk of families with t(1;2)(q42;q33) GTG, RHG, QFQ, FISH. Ginekol Pol 71: 1262–1272, 2000. Stasiewicz-Jarocka B, Haus O, Van Assche E, et al. Genetic counseling in carriers of reciprocal chromosomal translocations involving long arm of chromosome 16. Clin Genet 66: 189–207, 2004. Stefanou EG, Crocker M. A chromosome 21-derived minute marker in a mosaic trisomy 21 background: implications for risk assessments in marker chromosome cases. Am J Med Genet 127A : 191–193, 2004. Stefanou EG, Crocker M, Boon A , Stewart H. Cryptic mosaicism for monosomy 20 identified in renal tract cells. Clin Genet 70: 228–232, 2006. Stein QP, Boyle JG, Crotwell PL, et al. Prenatally diagnosed trisomy 20 mosaicism associated with arachnoid cyst of basal cistern. Prenat Diagn 28: 1169–1170, 2008. Steinbach P. Excess of mental retardation and/or congenital malformation in reciprocal translocations in man. Hum Genet 73: 379, 1986. Steinberg C, Zackai EH, Eunpu DL, Mennuti MT, Emanuel BS. Recurrence rate for de novo 21q21q translocation Down syndrome: a study of 112 families. Am J Med Genet 17: 523–530, 1984. Steinberg Warren N, Soukup S, King JL, Dignan PStJ. Prenatal diagnosis of trisomy 20 by chorionic villus sampling (CVS): a case report with long-term outcome. Prenat Diagn 21: 1111–1113, 2001. Stemkens D, Broekmans FJ, Kastrop PM, Hochstenbach R , Smith BG, Giltay JC. Variant Klinefelter syndrome 47,X,i(X)(q10),Y and normal 46,XY karyotype in monozygotic adult twins. Am J Med Genet 143A : 1906–1911, 2007. Stene J. Detection of higher recurrence risk for age-dependent chromosome abnormalities with an application to trisomy G1. (Down’s syndrome). Hum Hered 20: 112–122, 1970. Stene J. Comments on methods and results in: Sherman et al., “Segregation analysis of balanced pericentric inversions in pedigree data.” Clin Genet 30: 95–107, 1986. Stene J, Stene E, Mikkelsen M. Risk for chromosome abnormality at amniocentesis following a child with a non-inherited chromosome aberration. A European Collaborative Study on Prenatal Diagnoses 1981. Prenat Diagn 4: Spec No. 81–95, 1984. Stene J, Stengel-Rutkowski S. Genetic risks of familial reciprocal and Robertsonian translocation carriers. In: Daniel A, ed. The Cytogenetics of Mammalian
Autosomal Rearrangements. New York: AR Liss, 1988: 3–72. Stengel-Rutkowski S, Stene J, Gallano P. Risk estimates in balanced parental reciprocal translocations. Monographie des Annales de Génétique 1–147, 1988. Stephenson MD, Awartani KA , Robinson WP. Cytogenetic analysis of miscarriages from couples with recurrent miscarriage: a case-control study. Hum Reprod 17: 446–451, 2002. Stephenson WT, Poirier SM, Rubin L, Einhorn LH. Evaluation of reproductive capacity in germ cell tumor patients following treatment with cisplatin, etoposide, and bleomycin. J Clin Oncol 13: 2278–2280, 1995. Stern C, Pertile M, Norris H, Hale L, Baker HGW. Chromosome translocations in couples with in-vitro fertilization implantation failure. Hum Reprod 14: 2097–2101, 1999. Stetten G, Escallon CS, South ST, McMichael JL, Saul DO, Blakemore KJ. Reevaluating confined placental mosaicism. Am J Med Genet 131: 232–239, 2004. Stevens SJC, Smeets EEJGL, Blom E, et al. Identical cryptic partial monosomy 20pter and trisomy 20qter in three adult siblings due to a large maternal pericentric inversion: detection by MLPA and breakpoint mapping by SNP array analysis. Am J Med Genet 149A : 2226–2230, 2009. Stevenson DA , Brothman AR , Chen Z, BayrakToydemir P, Longo N. Paternal uniparental disomy of chromosome 14: confirmation of a clinicallyrecognizable phenotype. Am J Med Genet 130A : 88–91, 2004. Stewart DA , Bailey JD, Netley CT, Park E. Growth, development, and behavioral outcome from mid-adolescence to adulthood in subjects with chromosome aneuploidy: the Toronto Study. Birth Defects Orig Artic Ser 26: 131–188, 1990. Stipoljev F, Stanojevic M, Kurjak A . Familial pericentric inversion of chromosome 4: inv(4) (p16.1q12). Clin Genet 61: 386–388, 2002. Storto PD, Diehn TN, O’Malley DP, et al. Satellited chromosome 10 detected prenatally in a fetus and confirmed as mosaic in a parent. Prenat Diagn 19: 1088–1089, 1999. Strain L, Dean JCS, Hamilton MPR , Bonthron DT. A true hermaphrodite chimera resulting from embryo amalgamation after in vitro fertilization. N Engl J Med 338: 166–169, 1998. Strain L, Warner JP, Johnston T, Bonthron DT. A human parthenogenetic chimaera. Nat Genet 11: 164–169, 1995. Streuli I, Fraisse T, Ibecheole V, Moix I, Morris MA , de Ziegler D. Intermediate and premutation FMR1
REFERENCES
•
601
alleles in women with occult primary ovarian insufficiency. Fertil Steril 92: 464–470, 2009. Strigini P, Pierluigi M, Forni GL, et al. Effect of x-rays on chromosome 21 nondisjunction. Am J Med Genet Suppl 7: 155–159, 1990. Stumm M, Müsebeck J, Tönnies H, et al. Partial trisomy 9p12p21.3 with a normal phenotype. J Med Genet 39: 141–144, 2002. Stuppia L, Calabrese G, Borrelli P, et al. Loss of the SHOX gene associated with Leri-Weill dyschondrosteosis in a 45,X male. J Med Genet 36: 711–713, 1999. Stuppia L, Gatta V, Calabrese G, et al. A quarter of men with idiopathic oligo-azoospermia display chromosomal abnormalities and microdeletions of different types in interval 6 of Yq11. Hum Genet 102: 566–570, 1998. Sturtevant AH, Beadle GW. An Introduction in Genetics. New York: Dover, 1962. Sudha T, Gopinath PM. Homologous Robertsonian translocation (21q21q) and abortions. Hum Genet 85: 253–255, 1990. Sudik R , Jakubiczka S, Nawroth F, Gilberg E, Wieacker PF. Chimerism in a fertile woman with 46,XY karyotype and female phenotype. Hum Reprod 16: 56–58, 2001. Sugawara N, Tokunaga Y, Maeda M, Komaba R , Araki Y. A successful pregnancy outcome using frozen testicular sperm from a chimeric infertile male with a 46, XX/46, XY karyotype: case report. Hum Reprod 20: 147–148, 2005. Sullivan AK , Marcus M, Epstein MP, et al. Association of FMR1 repeat size with ovarian dysfunction. Hum Reprod 20: 402–412, 2005. Sun F, Oliver-Bonet M, Turek PJ, Ko E, Martin RH. Meiotic studies in an azoospermic human translocation (Y;1) carrier. Mol Hum Reprod 11: 361–364, 2005. Sung PL, Chang SP, Wen KC, et al. Small supernumerary marker chromosome originating from chromosome 10 associated with an apparently normal phenotype. Am J Med Genet 149A : 2768–2774, 2009. Surace C, Piazzolla S, Sirleto P, et al. Mild ring 17 syndrome shares common phenotypic features irrespective of the chromosomal breakpoints location. Clin Genet 76: 256–262, 2009. Surti U, Szulman AE, Wagner K , Leppert M, O’Brien SJ. Tetraploid partial hydatidiform moles: two cases with a triple paternal contribution and a 92,XXXY karyotype. Hum Genet 72: 15–21, 1986. Susanne GO, Sissel S, Ulla W, Charlotta G, Sonja OL. Pregnant women’s responses to information about an increased risk of carrying a baby with Down syndrome. Birth 33: 64–73, 2006.
602
•
REFERENCES
Susiarjo M, Hassold TJ, Freeman E, Hunt PA . Bisphenol A exposure in utero disrupts early oogenesis in the mouse. PLoS Genet 3: e5, 2007. Sutherland GR . Fragile sites and cancer breakpoints—the pessimistic view. Cancer Genet Cytogenet 31: 5–7, 1988. Sutherland GR . The detection of fragile sites on human chromosomes. In: Adolph KW, ed. Advanced Techniques in Chromosome Research. New York: Marcel Dekker, 1991: 323–327. Sutherland GR . Rare fragile sites. Cytogenet Genome Res 100: 77–84, 2003. Sutherland GR , Baker E. The clinical significance of fragile sites on human chromosomes. Clin Genet 58: 157–161, 2000. Sutherland GR , Callen DF, Gardner RJM. Paracentric inversions do not normally generate monocentric recombinant chromosomes. Am J Med Genet 59: 390–392, 1995. Sutherland GR , Carter RF. 46,XX/46,XX,r (2) (p25q37) mosaicism: clinical and cytogenetic studies. Ann Génét 21: 164–167, 1978. Sutherland GR , Gardiner AJ, Carter RF. Familial pericentric inversion of chromosome 19, inv(19) (p13q13) with a note on genetic counseling of pericentric inversion carriers. Clin Genet 10: 54–59, 1976. Sutherland GR , Hecht F. Fragile Sites on Human Chromosomes. New York: Oxford Monographs on Medical Genetics, Oxford University Press, 1985. Sutherland GR , Richards RI. Dynamic mutations on the move. J Med Genet 30: 978–981, 1993. Sutherland GR , Simmers RN. No statistical association between common fragile sites and nonrandom chromosome breakpoints in cancer cells. Cancer Genet Cytogenet 31: 9–15, 1988. Sutton EJ, McInerney-Leo A , Bondy CA , Gollust SE, King D, Biesecker B. Turner syndrome: four challenges across the lifespan. Am J Med Genet 139A : 57–66, 2005. Sutton EJ, Young J, McInerney-Leo A , Bondy CA , Gollust SE, Biesecker BB. Truth-telling and Turner Syndrome: the importance of diagnostic disclosure. J Pediatr 148: 102–107, 2006. Sutton VR , McAlister WH, Bertin TK , et al. Skeletal defects in paternal uniparental disomy for chromosome 14 are re-capitulated in the mouse model (paternal uniparental disomy 12). Hum Genet 113: 447–451, 2003. Sutton VR , Shaffer LG. Search for imprinted regions on chromosome 14: comparison of maternal and paternal UPD cases with cases of chromosome 14 deletion. Am J Med Genet 93: 381–387, 2000. Swerdlow AJ, Hermon C, Jacobs PA , et al. Mortality and cancer incidence in persons with numerical
sex chromosome abnormalities: a cohort study. Ann Hum Genet 65: 177–188, 2001. Swerdlow AJ, Jacobs PA , Marks A , et al. Fertility, reproductive outcomes, and health of offspring, of patients treated for Hodgkin’s disease: an investigation including chromosome examinations. Br J Cancer 74: 291–296, 1996. Swillen A , Devriendt K , Vantrappen G, et al. Familial deletions of chromosome 22q 11: the Leuven experience. Am J Med Genet 80: 531–532, 1998. Swillen A , Vogels A , Devriendt K , Fryns JP. Chromosome 22q11 deletion syndrome: update and review of the clinical features, cognitivebehavioral spectrum, and psychiatric complications. Am J Med Genet 97: 128–135, 2000. Swinkels MEM, Simons A , Smeets DF, et al. Clinical and cytogenetic characterization of 13 Dutch patients with deletion 9p syndrome: delineation of the critical region for a consensus phenotype. Am J Med Genet 146A : 1430–1438, 2008. Sybert VP. Phenotypic effects of mosaicism for a 47,XXX cell line in Turner syndrome. J Med Genet 39: 217–220, 2002. Sybert VP. Turner syndrome. In: Cassidy SB, Allanson JE, eds. Management of Genetic Syndromes. Hoboken, NJ: John Wiley & Sons, 2004. Sybert VP, Koeberl D, McGillivray B. Reply to Hsu—”Prenatal diagnosis of 45,X/46,XX .” Am J Hum Genet 58: 635–636, 1996. Syme RM, Martin RH. Meiotic segregation of a 21;22 Robertsonian translocation. Hum Reprod 7: 825–829, 1992. Tachdjian G, Aboura A , Benkhalifa M, et al. De novo interstitial direct duplication of Xq21.1q25 associated with skewed X-inactivation pattern. Am J Med Genet 131: 273–280, 2004. Tacheva DM, Vladimirov IK . Embryos, preembryos, and stem cells. Fertil Steril 78: 1354–1355, 2002. Tagaya M, Mizuno S, Hayakawa M, Yokotsuka T, Shimizu S, Fujimaki H. Recombination of a maternal pericentric inversion results in 22q13 deletion syndrome. Clin Dysmorphol 17: 19–21, 2008. Taine L, Goizet C, Wen ZQ, et al. Submicroscopic deletion of chromosome 16p13.3 in patients with Rubinstein-Taybi syndrome. Am J Med Genet 78: 267–270, 1998. Takano K , Lyons M, Moyes C, Jones J, Schwartz CE. Two percent of patients suspected of having Angelman syndrome have TCF4 mutations. Clin Genet 78: 282–288, 2010. Takeno SS, Corbani M, Andrade JA , Smith Mde A , Brunoni D, Melaragno MI. Duplication 4p and
deletion 4p (Wolf-Hirschhorn syndrome) due to complementary gametes from a 3:1 segregation of a maternal balanced t(4;13)(p16;q11) translocation. Am J Med Genet 129A : 180–183, 2004. Takeuchi A , Ehara H, Ohtani K , et al. Live birth prevalence of Down syndrome in Tottori, Japan, 1980–1999. Am J Med Genet 146A : 1381–1386, 2008. Tamagaki A , Shima M, Tomita R , et al. Segregation of a pure form of spastic paraplegia and NOR insertion into Xq11.2. Am J Med Genet 94: 5–8, 2000. Tan TY, Aftimos S, Worgan L, et al. Phenotypic expansion and further characterisation of the 17q21.31 microdeletion syndrome. J Med Genet 46: 480–489, 2009. Tan-Sindhunata G, Castedo S, Leegte B, et al. Molecular cytogenetic characterization of a small, familial supernumerary ring chromosome 7 associated with mental retardation and an abnormal phenotype. Am J Med Genet 92: 147–152, 2000. Tang W, Boyd BK , Hummel M, Wenger SL. Prenatal diagnosis of tetrasomy 9p. Am J Med Genet 126A : 328, 2004. Tao T, Del Valle A . Human oocyte and ovarian tissue cryopreservation and its application. J Assist Reprod Genet 25: 287–296, 2008. Tarani L, Lampariello S, Raguso G, et al. Pregnancy in patients with Turner’s syndrome: six new cases and review of literature. Gynecol Endocrinol 12: 83–87, 1998. Tatton-Brown K , Pilz DT, Örstavik KH, et al. 15q overgrowth syndrome: a newly recognized phenotype associated with overgrowth, learning difficulties, characteristic facial appearance, renal anomalies and increased dosage of distal chromosome 15q. Am J Med Genet 149A : 147–154, 2009. Taylor AMR . Chromosome instability syndromes. Best Pract Res Clin Haematol 14: 631–644, 2001. Tekin M, Jackson-Cook C, Buller A , et al. Fluorescence in situ hybridization detectable mosaicism for Angelman syndrome with biparental methylation. Am J Med Genet 95: 145–149, 2000. Telvi L, Lebbar A , Del Pino O, Barbet JP, Chaussain JL. 45,X/46,XY mosaicism: report of 27 cases. Pediatrics 104: 304–308, 1999. Tempest HG, Ko E, Chan P, Robaire B, Rademaker A , Martin RH. Sperm aneuploidy frequencies analysed before and after chemotherapy in testicular cancer and Hodgkin’s lymphoma patients. Hum Reprod 23: 251–258, 2008.
REFERENCES
•
603
Templado C, Donate A , Giraldo J, Bosch M, Estop A. Advanced age increases chromosome structural abnormalities in human spermatozoa. Eur J Hum Genet 19: 145–151, 2011. Temple IK , Gardner RJ, Robinson DO, et al. Further evidence for an imprinted gene for neonatal diabetes localised to chromosome 6q22-q23. Hum Mol Genet 5: 1117–1121, 1996. Temple IK , Shield JP. Transient neonatal diabetes, a disorder of imprinting. J Med Genet 39: 872–875, 2002. Ten SK , Chin YM, Noor PJ, Hassan K . Cytogenetic studies in women with primary amenorrhea. Singapore Med J 31: 355–359, 1990. Tennakoon J, Kandasamy Y, Alcock G, Koh TH. Edwards syndrome with double trisomy. Singapore Med J 49: e190–191, 2008. Terribas E, Bonache S, García-Arévalo M, et al. Changes in the expression profile of the meiosisinvolved mismatch repair (MMR) genes in impaired human spermatogenesis. J Androl 31: 346–357, 2010. Terzoli G, Lalatta F, Lobbiani A , Simoni G, Colucci G. Fertility in a 47,XXY patient: assessment of biological paternity by deoxyribonucleic acid fingerprinting. Fertil Steril 58: 821–822, 1992. Terzoli G, Rossella F, Biscaglia M, Simoni G. True fetal mosaicism revealed by a single abnormal colony in amniocyte culture. Prenat Diagn 10: 273–274, 1990. Teshima IE, Winsor EJT, Van Allen MI. Trisomy 18 and a constitutional maternal translocation (2;18). Am J Med Genet 43: 759–761, 1992. Teyssier M, Gaucherand P, Buenerd A . Prenatal diagnosis of a tetraploid fetus. Prenat Diagn 17: 474–478, 1997. Teyssier M, Moreau N. Familial transmission of deleted chromosome 22 [r(22)p0?] in two normal women. Ann Génét 28: 116–118, 1985. Teyssier M, Rafat A , Pugeat M. Case of (Y;1) familial translocation. Am J Med Genet 46: 339–340, 1993. Thangavelu M, Frolich G, Rogers D. Partial duplication 2p as the sole abnormality in two cases with anencephaly. Am J Med Genet 124A : 170–172, 2004. Tharapel AT, Elias S, Shulman LP, Seely L, Emerson DS, Simpson JL. Resorbed co-twin as an explanation for discrepant chorionic villus results: non-mosaic 47,XX,+16 in villi (direct and culture) with normal (46,XX) amniotic fluid and neonatal blood. Prenat Diagn 9: 467–472, 1989. Tharapel AT, Michaelis RC, Velagaleti GVN, et al. Chromosome duplications and deletions and their mechanisms of origin. Cytogenet Cell Genet 85: 285–290, 1999.
604
•
REFERENCES
Tharapel AT, Tharapel SA , Bannerman RM. Recurrent pregnancy losses and parental chromosome abnormalities: a review. Br J Obstet Gynaecol 92: 899–914, 1985. Tharapel SA , Lewandowski RC, Tharapel AT, Wilroy RS, Jr. Phenotype-karyotype correlation in patients trisomic for various segments of chromosome 13. J Med Genet 23: 310–315, 1986. Theilgaard A . Psychologic study of XYY and XXY men. Birth Defects Orig Artic Ser 22: 277–292, 1986. Theisen A , Rosenfeld JA , Farrell SA , et al. aCGH detects partial tetrasomy of 12p in blood from Pallister-Killian syndrome cases without invasive skin biopsy. Am J Med Genet 149A : 914–918, 2009. Therman E, Laxova R , Susman B. The critical region on the human Xq. Hum Genet 85: 455–461, 1990. Therman E, Susman B, Denniston C. The nonrandom participation of human acrocentric chromosomes in Robertsonian translocations. Ann Hum Genet 53: 49–65, 1989. Thienpont B, Béna F, Breckpot J, et al. Duplications of the critical Rubinstein-Taybi deletion region on chromosome 16p13.3 cause a novel recognisable syndrome. J Med Genet 47: 155–161, 2010. Tho SP, Jackson R , Kulharya AS, Reindollar RH, Layman LC, McDonough PG. Long-term follow-up and analysis of monozygotic twins concordant for 45,X/46,XY peripheral blood karyotype but discordant for phenotypic sex. Am J Med Genet 143A : 2616–2622, 2007. Thomas JA , Johnson J, Peterson Kraai TL, et al. Genetic and clinical characterization of patients with an interstitial duplication 15q11-q13, emphasizing behavioral phenotype and response to treatment. Am J Med Genet 119A :111–120, 2003a. Thomas NS, Ennis S, Sharp AJ, et al. Maternal sex chromosome non-disjunction: evidence for X chromosome-specific risk factors. Hum Mol Genet 10: 243–250, 2001. Thomas NS, Hassold TJ. Aberrant recombination and the origin of Klinefelter syndrome. Hum Reprod Update 9:309–317, 2003. Thomas NS, Maloney V, Bryant V, et al. Breakpoint mapping and haplotype analysis of three reciprocal translocations identify a novel recurrent translocation in two unrelated families: t(4;11) (p16.2;p15.4). Hum Genet 125: 181–188, 2009. Thomas NS, Morris JK , Baptista J, Ng BL, Crolla JA , Jacobs PA . De novo apparently balanced translocations in man are predominantly paternal in origin and associated with a significant increase in paternal age. J Med Genet 47: 112–115, 2010.
Thomas NS, Roberts SE, Browne CE. Estimate of the prevalence of chromosome 15q11-q13 duplications. Am J Med Genet 119A : 111–120, 2003b. Thompson NM, Gulley ML, Rogeness GA , et al. Neurobehavioral characteristics of CGG amplification status in fragile X females. Am J Med Genet 54: 378–383, 1994. Tiepolo L, Zuffardi O. Localization of factors controlling spermatogenesis in the nonfluorescent portion of the human Y chromosome long arm. Hum Genet 34: 119–124, 1976. Tihy F, Lemieux N, Lemyre E. Complex chromosome rearrangement and recombinant balanced translocation in a mother and a daughter with the same phenotypic abnormalities. Am J Med Genet 135: 317–319, 2005. Tihy F, Lemyre E, Lemieux N, Dallaire L. De novo dup(X)(q22.1q25) in a girl with an abnormal phenotype. Am J Med Genet 87: 302–305, 1999. Till M, Rafat A , Charrin C, Plauchu H, Germain D. Duplication of chromosome 11 centromere in fetal and maternal karyotypes: a new variant? Prenat Diagn 11: 481–482, 1991. Tillisch DL. Chromosome deletions. Looking beyond disability. Lancet 358(suppl): S10, 2001. Tinkle BT, Walker ME, Blough-Pfau RI, Saal HM, Hopkin RJ. Unexpected survival in a case of prenatally diagnosed non-mosaic trisomy 22: Clinical report and review of the natural history. Am J Med Genet 118A : 90–95, 2003. Tiranti V, Lamantea E, Uziel G, et al. Leigh syndrome transmitted by uniparental disomy of chromosome 9. J Med Genet 36: 927–928, 1999. Tischkowitz M, Easton DF, Ball J, Hodgson SV, Mathew CG. Cancer incidence in relatives of British Fanconi Anaemia patients. BMC Cancer 8: 257, 2008. Tischkowitz MD, Hodgson SV. Fanconi anaemia. J Med Genet 40: 1–10, 2003. Toledano-Alhadef H, Basel-Vanagaite L, Magal N, et al. Fragile-X carrier screening and the prevalence of premutation and full-mutation carriers in Israel. Am J Hum Genet 69: 351–360, 2001. Tomaselli S, Megiorni F, De Bernardo C, et al. Syndromic true hermaphroditism due to an R-spondin1 (RSPO1) homozygous mutation. Hum Mutat 29: 220–226, 2008. Tomkins DJ. Unstable familial translocations: A t(11;22)mat inherited as a t(11;15). Am J Hum Genet 33: 745–751, 1981. Tommerup N. Mendelian cytogenetics. Chromosome rearrangements associated with mendelian disorders. J Med Genet 30: 713–727, 1993.
Tonk V, Schultz RA , Christian SL, Kubota T, Ledbetter DH, Wilson GN. Robertsonian (15q;15q) translocation in a child with Angelman syndrome: evidence of uniparental disomy. Am J Med Genet 66: 426–428, 1996. Tonk VS, Jesurun CA , Morgan DL, Lockhart LH, Velagaleti GV. Molecular cytogenetic characterization of a recombinant chromosome rec(22)dup(22q)inv(22)(p13q12.2). Am J Med Genet 124A : 92–95, 2004. Tonk VS, Wyandt HE, Huang X , et al. Disease associated balanced chromosome rearrangements (DBCR): report of two new cases. Ann Génét 46: 37–43, 2003. Torfs CP, Christianson RE. Anomalies in Down syndrome individuals in a large population-based registry. Am J Med Genet 77: 431–438, 1998. Torfs CP, Christianson RE. Effect of maternal smoking and coffee consumption on the risk of having a recognized Down syndrome pregnancy. Am J Epidemiol 152: 1185–1191, 2000. Torisu H, Yamamoto T, Fujiwaki T, et al. Girl with monosomy 1p36 and Angelman syndrome due to unbalanced der(1) transmission of a maternal translocation t(1;15)(p36.3;q13.1). Am J Med Genet 131: 94–98, 2004. Torniero C, Dalla Bernardina B, Novara F, et al. Dysmorphic features, simplified gyral pattern and 7q11.23 duplication reciprocal to the WilliamsBeuren deletion. Eur J Hum Genet 16: 880–887, 2008. Torrisi L, Sangiorgi E, Russo L, Gurrieri F. Rearrangements of chromosome 15 in epilepsy. Am J Med Genet 106: 125–128, 2001. Torun D, Bahçe M, Alanbay I, Guran S, Baser I. Prenatal diagnosis of Cri-du chat syndrome following high maternal serum human chorionic gonodotrophin and choroid plexus cysts. Prenat Diagn 29: 536–537, 2009. Tosca L, Brisset S, Petit FM, et al. Recurrent 70.8 Mb 4q22.2q32.3 duplication due to ovarian germinal mosaicism. Eur J Hum Genet 18: 882–888, 2010. Tóth A , Gaál M, László J. Familial pericentric inversion of the Y chromosome. Ann Génét 27: 60–61, 1984. Touliatou V, Mavrou A , Kolialexi A , Kanavakis E, Kitsiou-Tzeli S. Saethre-Chotzen syndrome with severe developmental delay associated with deletion of chromosomic region 7p15 → pter. Genet Couns 18: 295–301, 2007. Towner DR , Shaffer LG, Yang SP, Walgenbach DD. Confined placental mosaicism for trisomy 14 and maternal uniparental disomy in association with elevated second trimester maternal serum human chorionic gonadotrophin and third trimester fetal
REFERENCES
•
605
growth restriction. Prenat Diagn 21: 395–398, 2001. Tranebjaerg L, Petersen A , Hove K , Rehder H, Mikkelsen M. Clinical and cytogenetic studies in a large (4;8) translocation family with pre- and postnatal Wolf syndrome. Ann Génét 27: 224–229, 1984. Trask BJ. Human cytogenetics: 46 chromosomes, 46 years and counting. Nat Rev Genet 3: 769–778, 2002. Traversa MV, Carey L, Leigh D. A molecular strategy for routine preimplantation genetic diagnosis in both reciprocal and Robertsonian translocation carriers. Mol Hum Reprod 16: 329–337, 2010. Traylor RN, Fan Z, Hudson B, et al. Microdeletion of 6q16.1 encompassing EPHA7 in a child with mild neurological abnormalities and dysmorphic features: case report. Mol Cytogenet 2: 17, 2009. Treetampinich C, O’Connor AE, MacLachlan V, Groome NP, de Kretser DM. Maternal serum inhibin A concentrations in early pregnancy after IVF and embryo transfer reflect the corpus luteum contribution and pregnancy outcome. Hum Reprod 15: 2028–2032, 2000. Treff NR , Levy B, Su J, Northrop LE, Tao X , Scott RT. SNP microarray based 24 chromosome aneuploidy screening is significantly more consistent than FISH. Mol Hum Reprod 16: 583–589, 2010a. Treff NR , Northrop LE, Kasabwala K , Su J, Levy B, Scott RT. Single nucleotide polymorphism microarray-based concurrent screening of 24-chromosome aneuploidy and unbalanced translocations in preimplantation human embryos. Fertil Steril 95: 1606-1612.e1-2, 2011a. Treff NR , Su J, Tao X , Katz-Jaffe M, Schoolcraft W, Scott R . Characterizing the nature of meiotic errors: PSSC is the principal meiotic error in human oocytes and may correct during the second meiotic division and result in the birth of a healthy infant. Fertil Steril 92: S25, 2009a. Treff NR , Su J, Tao X , Levy B, Scott RT. Accurate single cell 24 chromosome aneuploidy screening using whole genome amplification and single nucleotide polymorphism microarrays. Fertil Steril 94: 2017–2021, 2010b. Treff N, Su J, Tao X , Miller K , Scott R . First IVF babies born after rapid 24 chromosome embryo aneuploidy screening and fresh embryo transfer. Fertil Steril 92: S49, 2009b. Treff NR , Tao X, Schillings WJ, Bergh, PA, Scott RT, Levy B. Use of single nucleotide polymorphism microarrays to distinguish between balanced and normal chromosomes in embryos from a translocation carrier. Fertil Steril 96: e58–65, 2011b.
606
•
REFERENCES
Troche V, Hernandez E. Neoplasia arising in dysgenetic gonads. Obstet Gynecol Surv 41: 74–79, 1986. Trujillo-Tiebas MJ, González-González C, LordaSánchez I, Querejeta ME, Ayuso C, Ramos C. Prenatal diagnosis of 46, XX male fetus. J Assist Reprod Genet 23: 253–254, 2006. Tsai AC, Fine CA , Yang M, Walton CS, Beischel L, Johnson JP. De novo isodicentric X chromosome: 46,X,idic(X)(q24), and summary of literature. Am J Med Genet 140: 923–930, 2006. Tsai AC, Gibby T, Beischel L, McGavran L, Johnson JP. A child with Angelman syndrome and trisomy 13 findings due to associated paternal UPD 15 and segmental UPD 13. Am J Med Genet 126A : 208–212, 2004. Tseng LH, Chuang SM, Lee TY, Ko TM. Recurrent Down’s syndrome due to maternal ovarian trisomy 21 mosaicism. Arch Gynecol Obstet 255: 213–216, 1994. Tsezou A , Hadjiathanasiou C, Gourgiotis D, et al. Molecular genetics of Turner syndrome: correlation with clinical phenotype and response to growth hormone therapy. Clin Genet 56: 441–446, 1999. Tsien F, Fiala ES, Youn B, et al. Prolonged culture of normal chorionic villus cells yields ICF syndrome-like chromatin decondensation and rearrangements. Cytogenet Genome Res 98: 13–21, 2002. Tsilchorozidou T, Menko FH, Lalloo F, et al. Constitutional rearrangements of chromosome 22 as a cause of neurofibromatosis 2. J Med Genet 41: 529–534, 2004. Tucker ME, Garringer HJ, Weaver DD. Phenotypic spectrum of mosaic trisomy 18: two new patients, a literature review, and counseling issues. Am J Med Genet 143: 505–517, 2007. Tuck-Muller CM, Chen H, Martínez JE, et al. Isodicentric Y chromosome: cytogenetic, molecular and clinical studies and review of the literature. Hum Genet 96: 119–129, 1995. Tul N, Novak-Antolič Z. Serum PAPP-A levels at 10–14 weeks of gestation are altered in women after assisted conception. Prenat Diagn 26: 1206–1211, 2006. Tümer Z, Wolff D, Silahtaroglu AN, Ørum A , Brøndum-Nielsen K . Characterization of a supernumerary small marker X chromosome in two females with similar phenotypes. Am J Med Genet 76: 45–50, 1998. Tupler R , Barbierato L, Larizza D, Sampaolo P, Piovella F, Maraschio P. Balanced autosomal translocations and ovarian dysgenesis. Hum Genet 94: 171–176, 1994.
Turleau C. Monosomy 18p. Orphanet J Rare Dis 3: 4, 2008. Turner C, Dennis NR , Skuse DH, Jacobs PA . Seven ring (X) chromosomes lacking the XIST locus, six with an unexpectedly mild phenotype. Hum Genet 106: 93–100, 2000. Turner G. Finding genes on the X chromosome by which homo may have become sapiens. Am J Hum Genet 58: 1109–1110, 1996. Turner JMA . Meiotic sex chromosome inactivation. Development 134: 1823–1831, 2007. Turnpenny PD, Pigott RW. Deletion 22q11 syndrome: acknowledging a lost eponym as we say farewell to an acronym. J Med Genet 38: 271–273, 2001. Twisk M, Mastenbroek S, Hoek A , et al. No beneficial effect of preimplantation genetic screening in women of advanced maternal age with a high risk for embryonic aneuploidy. Hum Reprod 23: 2813–2817, 2008. Tyler CT, Rice GM, Grady M, Raca G. Mild clinical presentation in a child with prenatally diagnosed 45,X/47,XX,+18 mosaicism. Am J Med Genet 149A : 2588–2592, 2009. Tyreman M, Abbott KM, Willatt LR , et al. High resolution array analysis: diagnosing pregnancies with abnormal ultrasound findings. J Med Genet 46: 531–541, 2009. Tyshchenko N, Lurie I, Schinzel A . Chromosomal map of human brain malformations Hum Genet 124: 73–80, 2009. Tzoufi M, Kanioglou C, Dasoula A , et al. Mosaic trisomy r(14) associated with epilepsy and mental retardation. J Child Neurol 22: 869–873, 2007. Tzschach A , Chen W, Erdogan F, et al. Characterization of interstitial Xp duplications in two families by tiling path array CGH. Am J Med Genet 146A : 197–203, 2008. Uchida IA , Freeman VCP. Trisomy 21 Down syndrome. II. Structural chromosome rearrangements in the parents. Hum Genet 72: 118–122, 1986. Uehara S, Funato T, Yaegashi N, et al. SRY mutation and tumor formation on the gonads of XY pure gonadal dysgenesis patients. Cancer Genet Cytogenet 113: 78–84, 1999a. Uehara S, Nata M, Obara Y, Niinuma T, Funato T, Yajima A . A Turner syndrome woman with a ring X chromosome [45,X/46,X,r(X)(p22.3q27)] whose child also had a ring X chromosome. Fertil Steril 67: 576–579, 1997. Uehara S, Tamura M, Nata M, et al. Complete androgen insensitivity in a 47,XXY patient with uniparental disomy for the X chromosome. Am J Med Genet 86: 107–111, 1999b.
Uehara S, Yaegashi N, Maeda T, et al. Risk of recurrence of fetal chromosomal aberrations: analysis of trisomy 21, trisomy 18, trisomy 13, and 45,X in 1,076 Japanese mothers. J Obstet Gynaecol Res 25: 373–379, 1999c. Uematsu A , Yorifuji T, Muroi J, et al. Parental origin of normal X chromosomes in Turner syndrome patients with various karyotypes: implications for the mechanism leading to generation of a 45,X karyotype. Am J Med Genet 111: 134–139, 2002. Uhrig S, Schlembach D, Waldispuehl-Geigl J, et al. Impact of array comparative genomic hybridization-derived information on genetic counseling demonstrated by prenatal diagnosis of the TAR (thrombocytopenia-absent-radius) syndrome-associated microdeletion 1q21.1. Am J Hum Genet 81: 866–868, 2007. Ulm JE. Recurrent trisomies: chance or inherited predisposition? J Genet Counsel 8: 109–117, 1999. Urban M, Bommer C, Tennstedt C, et al. Ring chromosome 6 in three fetuses: case reports, literature review, and implications for prenatal diagnosis. Am J Med Genet 108: 97–104, 2002. Urioste M, Martínez-Frías ML, Bermejo E, et al. Short rib-polydactyly syndrome and pericentric inversion of chromosome 4. Am J Med Genet 49: 94–97, 1994a. Urioste M, Visedo G, Sanchís A , et al. Dynamic mosaicism involving an unstable supernumerary der(22) chromosome in cat eye syndrome. Am J Med Genet 49: 77–82, 1994b. Uroz L, Rajmil O, Templado C. Premature separation of sister chromatids in human male meiosis. Hum Reprod 23: 982–987, 2008. Utermann B, Riegel M, Leistritz D, et al. Pre- and postnatal findings in trisomy 17 mosaicism. Am J Med Genet 140: 1628–1636, 2006. Valle L, Cascón A , Melchor L, et al. About the origin and development of hereditary conventional renal cell carcinoma in a four-generation t(3;8) (p14.1;q24.23) family. Eur J Hum Genet 13: 570–578, 2005. Van Assche E, Staessen C, Vegetti W, et al. Preimplantation genetic diagnosis and sperm analysis by fluorescence in-situ hybridization for the most common reciprocal translocation t(11;22). Mol Hum Reprod 5: 682–690, 1999. Van Bon BWM, Koolen DA , Brueton L, et al. The 2q23.1 microdeletion syndrome: clinical and behavioural phenotype. Eur J Hum Genet 18: 163–170, 2010. Van Bon BWM, Mefford HC, Menten B, et al. Further delineation of the 15q13 microdeletion and
REFERENCES
•
607
duplication syndromes: a clinical spectrum varying from non-pathogenic to a severe outcome. J Med Genet 46: 511–523, 2009. Van Buggenhout G, Fryns JP. Angelman syndrome (AS, MIM 105830). Eur J Hum Genet 17: 1367–1373, 2009. Van Buggenhout GJCM, Pijkels E, Holvoet M, Schaap C, Hamel BCJ, Fryns JP. Cri du chat syndrome: changing phenotype in older patients. Am J Med Genet 90: 203–215, 2000. Van de Laar I, Rabelink G, Hochstenbach R , Tuerlings J, Hoogeboom J, Giltay J. Diploid/triploid mosaicism in dysmorphic patients. Clin Genet 62: 376–382, 2002. Van de Leemput J, Chandran J, Knight MA , et al. Deletion at ITPR1 underlies ataxia in mice and spinocerebellar ataxia 15 in humans. PLoS Genet 3: e108, 2007. Van de Velde H, De Vos A , Sermon K , et al. Embryo implantation after biopsy of one or two cells from cleavage-stage embryos with a view to preimplantation genetic diagnosis. Prenat Diagn 20: 1030–1037, 2000. Van den Berg C, Van Opstal D, Brandenburg H, Los FJ. Case of 45,X/46,XY mosaicism with non-mosaic discordance between short-term villi (45,X) and cultured villi (46,XY). Am J Med Genet 93: 230–233, 2000. Van den Berg C, Van Opstal D, Polak-Knook J, Galjaard RJ. (Potential) false-negative diagnoses in chorionic villi and a review of the literature. Prenat Diagn 26: 401–408, 2006. Van den Berg DJ, Francke U. Roberts syndrome: a review of 100 cases and a new rating system for severity. Am J Med Genet 47: 1104–1123, 1993. Van den Berg IM, Laven JSE, Stevens M, et al. X chromosome inactivation is initiated in human preimplantation embryos. Am J Hum Genet 84: 771–779, 2009. Van den Veyver IB, Patel A , Shaw CA , et al. Clinical use of array comparative genomic hybridization (aCGH) for prenatal diagnosis in 300 cases. Prenat Diagn 29: 29–39, 2009. Van der Aa N, Rooms L, Vandeweyer G, et al. Fourteen new cases contribute to the characterization of the 7q11.23 microduplication syndrome. Eur J Med Genet 52: 94–100, 2009. Van der Burgt CJAM, Merkx GFM, Janssen AH, Mulder JC, Suijkerbuijk RF, Smeets DFCM. Partial trisomy for 5q and monosomy for 12p in a liveborn child as a result of a complex five breakpoint chromosome rearrangement in a parent. J Med Genet 29: 739–741, 1992. Van der Lelij P, Chrzanowska KH, Godthelp BC, et al. Warsaw breakage syndrome, a cohesinopathy
608
•
REFERENCES
associated with mutations in the XPD helicase family member DDX11/ChlR1. Am J Hum Genet 86: 262–266, 2010. Van der Luijt RB, Tops CMJ, Khan PM, et al. Molecular, cytogenetic, and phenotypic studies of a constitutional reciprocal translocation t(5;10) (q22;q25) responsible for familial adenomatous polyposis in a Dutch pedigree. Genes Chromosomes Cancer 13: 192–202, 1995. Van der Veken LT, Dieleman MMJ, Douben H, et al. Low grade mosaic for a complex supernumerary ring chromosome 18 in an adult patient with multiple congenital anomalies. Mol Cytogenet 3: 13, 2010. Van der Ven K , Peschka B, Montag M, Lange R , Schwanitz G, van der Ven HH. Increased frequency of congenital chromosomal aberrations in female partners of couples undergoing intracytoplasmic sperm injection. Hum Reprod 13: 48–54, 1998. Van der Zwaag B, Staal WG, Hochstenbach R , et al. A co-segregating microduplication of chromosome 15q11.2 pinpoints two risk genes for autism spectrum disorder. Am J Med Genet 153B: 960–966, 2010. Van Dyke DL, Weiss L, Roberson JR , Babu VR . The frequency and mutation rate of balanced autosomal rearrangements in man estimated from prenatal genetic studies for advanced maternal age. Am J Hum Genet 35: 301–308, 1983. Van Esch H, Groenen P, Fryns JP, Van de Ven W, Devriendt K . The phenotypic spectrum of the 10p deletion syndrome versus the classical DiGeorge syndrome. Genet Couns 10: 59–65, 1999. Van Esch H, Groenen P, Nesbit MA , et al. GATA3 haplo-insufficiency causes human HDR syndrome. Nature 406: 419–422, 2000. Van Haelst MM, Van Opstal D, Lindhout D, Los FJ. Management of prenatally detected trisomy 8 mosaicism. Prenat Diagn 21: 1075–1078, 2001. Van Hemel JO, Eussen HJ. Interchromosomal insertions. Identification of five cases and a review. Hum Genet 107: 415–432, 2000. Van Hummelen P, Lowe XR , Wyrobek AJ. Simultaneous detection of structural and numerical chromosome abnormalities in sperm of healthy men by multicolor fluorescence in situ hybridization. Hum Genet 98: 608–615, 1996. Van Hummelen P, Manchester D, Lowe X , Wyrobek AJ. Meiotic segregation, recombination, and gamete aneuploidy assessed in a t(1;10) (p22.1;q22.3) reciprocal translocation carrier by three- and four-probe multicolor FISH in sperm. Am J Hum Genet 61: 651–659, 1997.
Van Karnebeek CDM, Hennekam RCM. Associations between chromosomal anomalies and congenital heart defects: a database search. Am J Med Genet 84: 158–166, 1999. Van Kogelenberg M, Ghedia S, McGillivray G, et al. Periventricular heterotopia in common microdeletion syndromes. Mol Syndromol 1: 35–41, 2010. Van Opstal D, van den Berg C, Galjaard RJH, Los FJ. Follow-up investigations in uncultured amniotic fluid cells after uncertain cytogenetic results. Prenat Diagn 21: 75–80, 2001. Van Ravenswaaij-Arts CMA , Kleefstra T. Emerging microdeletion and microduplication syndromes; the counseling paradigm. Eur J Med Genet 52: 75–76, 2009. Van Roy N, Vandesompele J, Berx G, et al. Localization of the 17q breakpoint of a constitutional 1;17 translocation in a patient with neuroblastoma within a 25-kb segment located between the ACCN1 and TLK2 genes and near the distal breakpoints of two microdeletions in neurofibromatosis type 1 patients. Genes Chromosomes Cancer 35: 113–120, 2002. Van Silfhout AT, van den Akker PC, Dijkhuizen T, et al. Split hand/foot malformation due to chromosome 7q aberrations(SHFM1): additional support for functional haploinsufficiency as the causative mechanism. Eur J Hum Genet 17: 1432–1438, 2009. Van Zutven LJCM, van Bever Y, Van Nieuwland CCM, et al. Interstitial 11q deletion derived from a maternal ins(4;11)(p14;q24.2q25): a patient report and review. Am J Med Genet 149A : 1468–1475, 2009. Vandeweyer G, Kooy RF. Balanced translocations in mental retardation. Hum Genet 126: 133–147, 2009. Vanneste E, Voet T, Le Caignec C, et al. Chromosome instability is common in human cleavage-stage embryos. Nat Med 15: 577–583, 2009a. Vanneste E, Voet T, Melotte C, et al. What next for preimplantation genetic screening? High mitotic chromosome instability rate provides the biological basis for the low success rate. Hum Reprod 24: 2679–2682, 2009b. Varela M, Shapira E, Hyman DB. Ullrich-Turner syndrome in mother and daughter: prenatal diagnosis of a 46,X,del(X)(p21) offspring from a 45,X mother with low-level mosaicism for the del(X)(p21) in one ovary. Am J Med Genet 39: 411–412, 1991. Varela MC, Simões-Sato AY, Kim CA , Bertola DR , De Castro CI, Koiffmann CP. A new case of interstitial 6q16.2 deletion in a patient with Prader-Willi-like
phenotype and investigation of SIM1 gene deletion in 87 patients with syndromic obesity. Eur J Med Genet 49: 298–305, 2006. Varley JM. Patterns of silver staining of human chromosomes. Chromosoma 61: 207–214, 1977. Vauhkonen AE, Sankila EM, Simola KOJ, de la Chapelle A . Segregation and fertility analysis in an autosomal reciprocal translocation, t(1;8) (q41;q23.1). Am J Hum Genet 37: 533–542, 1985. Vázquez-Cárdenas A , Vásquez-Velásquez AI, Barros-Núñez P, Mantilla-Capacho J, Rocchi M, Rivera H. Familial whole-arm translocations (1;19), (9;13), and (12;21): a review of 101 constitutional exchanges. J Appl Genet 48: 261–268, 2007. Veenma DCM, Eussen HJ, Govaerts LCP, et al. Phenotype-genotype correlation in a familial IGF1R microdeletion case. J Med Genet 47: 492–498, 2010. Vega H, Trainer AH, Gordillo M, et al. Phenotypic variability in 49 cases of ESCO2 mutations, including novel missense and codon deletion in the acetyltransferase domain, correlates with ESCO2 expression and establishes the clinical criteria for Roberts syndrome. J Med Genet 47: 30–37, 2010. Vegetti W, Van Assche E, Frias A , et al. Correlation between semen parameters and sperm aneuploidy rates investigated by fluorescence in-situ hybridization in infertile men. Hum Reprod 15: 351–365, 2000. Velagaleti GVN, Lockhart LH, Schmalstieg FC, Goldman AS. Trisomy 4 pter-q12 and monosomy of chromosome 13 pter-q12 in a male with deficiency of all blood lymphocyte populations. Am J Med Genet 102: 139–145, 2001. Veld PA , Weber RF, Los FJ, et al. Two cases of Robertsonian translocations in oligozoospermic males and their consequences for pregnancies induced by intracytoplasmic sperm injection. Hum Reprod 12: 1642–1644, 1997. Velinov M, Dolzhanskaya N. Clavicular pseudoarthrosis, anomalous coronary artery and extra crease of the fifth finger-previously unreported features in individuals with class II 1q21.1 microdeletions. Eur J Med Genet 53: 213–216, 2010. Velissariou V, Antoniadi T, Gyftodimou J, et al. Maternal uniparental isodisomy 20 in a foetus with trisomy 20 mosaicism: clinical, cytogenetic and molecular analysis. Eur J Hum Genet 10: 694–698, 2002. Veltman JA , Brunner HG. Understanding variable expressivity in microdeletion syndromes. Nat Genet 42: 192–193, 2010.
REFERENCES
•
609
Vendola C, Canfield M, Daiger SP, et al. Survival of Texas infants born with trisomies 21, 18, and 13. Am J Med Genet 152A : 360–366, 2010. Veneman TF, Beverstock GC, Exalto N, Mollevanger P. Premature menopause because of an inherited deletion in the long arm of the X-chromosome. Fertil Steril 55: 631–633, 1991. Venter PA , Dawson B, Du Toit JL, et al. A familial paracentric inversion: a short review of the current status. Hum Genet 67: 121–125, 1984. Verghese S, Newlin A , Miller M, Burton BK . Mosaic trisomy 7 in a patient with pigmentary abnormalities. Am J Med Genet 87: 371–374, 1999. Verhoeven WM, Tuinier S, Curfs LM. Prader-Willi syndrome: the psychopathological phenotype in uniparental disomy. J Med Genet 40: e112, 2003. Verlinsky Y, Cieslak J, Ivakhnenko V, et al. Prevention of age-related aneuploidies by polar body testing of oocytes. J Assist Reprod Genet 16: 165–169, 1999. Verlinsky Y, Cieslak J, Kuliev A . Similar frequencies of meiosis I & II aneuploidies in IVF patients of advanced maternal age. Ann Génét 44: s124, 2001a. Verlinsky Y, Rechitsky S, Schoolcraft W, Strom C, Kuliev A . Preimplantation diagnosis for Fanconi anemia combined with HLA matching. J Am Med Assoc 285: 3130–3133, 2001b. Verloes A , Gillerot Y, Van Maldergem L, et al. Major decrease in the incidence of trisomy 21 at birth in south Belgium: mass impact of triple test? Eur J Hum Genet 9: 1–4, 2001. Verloes A , Lesenfants S, Jamar M, Dideberg V, Herens C. GOMBO syndrome: another “pseudorecessive” disorder due to a cryptic translocation. Am J Med Genet 95: 185–186, 2000. Verma RS, Rodriguez J, Dosik H. The clinical significance of pericentric inversion of the human Y chromosome: a rare “third” type of heteromorphism. J Hered 73: 236–238, 1982. Vermeesch JR , Baten E, Fryns JP, Devriendt K . Ring syndrome caused by ring chromosome 7 without loss of subtelomeric sequences. Clin Genet 62: 415–417, 2002. Vermeesch JR , Duhamel H, Petit P, Falzetti D, Fryns JP, Marynen P. Multiple small accessory marker chromosomes from different centromeric origin in a moderately mentally retarded male. Hum Genet 105: 611–618, 1999. Verp MS, Bombard AT, Simpson JL, Elias S. Parental decision following prenatal diagnosis of fetal chromosome abnormality. Am J Med Genet 29: 613–622, 1988. Verp MS, Simpson JL. Abnormal sexual differentiation and neoplasia. Cancer Genet Cytogenet 25: 191–218, 1987.
610
•
REFERENCES
Vialard F, Hammoud I, Molina-Gomes D, et al. Gamete cytogenetic study in couples with implantation failure: aneuploidy rate is increased in both couple members. J Assist Reprod Genet 25: 539–545, 2008. Vialard F, Nouchy M, Malan V, Taillemite JL, Selva J, Portnoï MF. Whole-arm translocations between chromosome 1 and acrocentric G chromosomes are associated with a poor prognosis for spermatogenesis: two new cases and review of the literature. Fertil Steril 86: 1001 e1–5, 2006. Vičić A , Roje D, Strinić T, Stipoljev F. Trisomy 1 in an early pregnancy failure. Am J Med Genet 146A : 2439–2441, 2008. Vickers S, Dahlitz M, Hardy C, Kilpatrick M, Webb T. A male with a de novo translocation involving loss of 15q11q13 material and Prader-Willi syndrome. J Med Genet 31: 478–481, 1994. Vignoli A , Canevini MP, Darra F, et al. Ring chromosome 20 syndrome: a link between epilepsy onset and neuropsychological impairment in three children. Epilepsia 50: 2420–2427, 2009. Vinci G, Brauner R , Tar A , et al. Mutations in the TSPYL1 gene associated with 46,XY disorder of sex development and male infertility. Fertil Steril 92: 1347–1350, 2009. Vinci G, Chantot-Bastaraud S, El Houate B, Lortat-Jacob S, Brauner R , McElreavey K . Association of deletion 9p, 46,XY gonadal dysgenesis and autistic spectrum disorder. Mol Hum Reprod 13: 685–689, 2007. Visootsak J, Graham JM, Jr. Klinefelter syndrome and other sex chromosomal aneuploidies. Orphanet J Rare Dis 1: 42, 2006. Vissers LELM, Bhatt SS, Janssen IM, et al. Rare pathogenic microdeletions and tandem duplications are microhomology-mediated and stimulated by local genomic architecture. Hum Mol Genet 18: 3579–3593, 2009. Vissers LELM, van Ravenswaaij CMA , Admiraal R , et al. Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nat Genet 36: 955–957, 2004. Vockley J, Inserra JA , Breg WR , Yang-Feng TL. “Pseudomosaicism” for 4p- in amniotic fluid cell culture proven to be true mosaicism after birth. Am J Med Genet 39: 81–83, 1991. Vogels A , Devriendt K , Vermeesch JR , et al. Cryptic translocation t(5;18) in familial mental retardation. Ann Génét 43: 117–123, 2000. Vogt J, Ryan E, Tischkowitz MD, Reardon W, Brueton LA . The tale of a nail sign in chromosome 4q34 deletion syndrome. Clin Dysmorphol 15: 127–132, 2006.
Voiculescu I, Barbi G, Wolff G, Steinbach P, Back E, Schempp W. Familial pericentric inversion of chromosome 12. Hum Genet 72: 320–322, 1986. Von der Lippe C, Rustada C, Heimdala K , Rødningena OK. 15q11.2 microdeletion—Seven new patients with delayed development and/or behavioural problems. Eur J Med Genet 54: 357-360, 2011. Voss R , Ben-Simon E, Avital A , et al. Isodisomy of chromosome 7 in a patient with cystic fibrosis: could uniparental disomy be common in humans? Am J Hum Genet 45: 373–380, 1989. Voullaire L, Collins V, Callaghan T, McBain J, Williamson R , Wilton L. High incidence of complex chromosome abnormality in cleavage embryos from patients with repeated implantation failure. Fertil Steril 87: 1053–1058, 2007. Voullaire L, Gardner RJM, Vaux C, Robertson A , Oertel R , Slater H. Chromosomal duplication of band 10p14 segregating through four generations. J Med Genet 37: 233–237, 2000a. Voullaire L, Saffery R , Earle E, et al. Mosaic inv dup(8p) marker chromosome with stable neocentromere suggests neocentromerization is a post-zygotic event. Am J Med Genet 102: 86–94, 2001. Voullaire LE, Slater HR , Petrovic V, Choo KHA . A functional marker centromere with no detectable alpha-satellite, satellite III, or CENP-B protein: activation of a latent centromere? Am J Hum Genet 52: 1153–1163, 1993. Voullaire L, Slater H, Williamson R , Wilton L. Chromosome analysis of blastomeres from human embryos by using comparative genomic hybridization. Hum Genet 106: 210–217, 2000b. Voullaire L, Wilton L, McBain J, Callaghan T, Williamson R . Chromosome abnormalities identified by comparative genomic hybridization in embryos from women with repeated implantation failure. Mol Hum Reprod 8: 1035–1041, 2002. Vozdova M, Horinova V, Wernerova V, et al. 2011. Der(4)t(Y;4): three-generation transmission and sperm meiotic segregation analysis. Am J Med Genet 155A : 1157–1161, 2011. Wagner T, Wirth J, Meyer J, et al. Autosomal sex reversal and campomelic dysplasia are caused by mutations in and around the SRY-related gene SOX9. Cell 79: 1111–1120, 1994. Wagstaff J, Hemann M. A familial “balanced” 3;9 translocation with cryptic 8q insertion leading to deletion and duplication of 9p23 loci in siblings. Am J Hum Genet 56: 302–309, 1995. Wahab MA , Nickless EM, Najar-M’Kacher R , Parmentier C, Podd JV, Rowland RE. Elevated
chromosome translocation frequencies in New Zealand nuclear test veterans. Cytogenet Genome Res 121: 79–87, 2008. Wakeling EL, Abu-Amero S, Alders M, et al. Epigenotype-phenotype correlations in SilverRussell syndrome. J Med Genet 47: 760–768, 2010. Wakui K , Gregato G, Ballif BC, et al. Construction of a natural panel of 11p11.2 deletions and further delineation of the critical region involved in Potocki-Shaffer syndrome. Eur J Hum Genet 13: 528–540, 2005. Walczak C, Enders H, Grissinger K , Dufke A . Retrospective diagnosis of trisomy 15 in formalinfixed, paraffin-embedded placental tissue in a newborn girl with Prader-Willi syndrome. Prenat Diagn 20: 914–916, 2000. Walczak-Sztulpa J, Wisniewska M, Latos-Bielenska A , et al. Chromosome deletions in 13q33–34: report of four patients and review of the literature. Am J Med Genet 146: 337–342, 2008. Wald NJ, Huttly WJ, Rudnicka AR . Prenatal screening for Down syndrome: the problem of recurrent false-positives. Prenat Diagn 24: 389–392, 2004. Wald NJ, Watt HC, Hackshaw AK . Integrated screening for Down’s syndrome on the basis of tests performed during the first and second trimesters. N Engl J Med 341: 461–467, 1999. Walker AP, Bocian M. Partial duplication 8q12→q21.2 in two sibs with maternally derived insertional and reciprocal translocations: case reports and review of partial duplications of chromosome 8. Am J Med Genet 27: 3–22, 1987. Walkinshaw SA . Fetal choroid plexus cysts: are we there yet? Prenat Diagn 20: 657–662, 2000. Wallace BMN, Hultén MA . Triple chromosome synapsis in oocytes from a human foetus with trisomy 21. Ann Hum Genet 47: 271–276, 1983. Wallace WHB, Kelsey TW. Human ovarian reserve from conception to the menopause. PLoS One 5: e8772, 2010. Wallerstein R , Anderson CE, Hay B, et al. Submicroscopic deletions at 16p13.3 in Rubinstein-Taybi syndrome: frequency and clinical manifestations in a North American population. J Med Genet 34: 203–206, 1997. Wallerstein R , Oh T, Durcan J, Abdelhak Y, Clachko M, Aviv H. Outcome of prenatally diagnosed trisomy 6 mosaicism. Prenat Diagn 22: 722–724, 2002. Wallerstein R , Strmen V, Durcan J, et al. Factors in decision making following genetic counseling for pre-natal diagnosis of de novo chromosomal rearrangements. Clin Genet 69: 497–503, 2006. Wallerstein R , Twersky S, Layman P, et al. Long term follow-up of developmental delay in a child with
REFERENCES
•
611
prenatally-diagnosed trisomy 20 mosaicism. Am J Med Genet 137: 94–97, 2005. Wallerstein R , Yu MT, Neu RL, et al. Common trisomy mosaicism diagnosed in amniocytes involving chromosomes 13, 18, 20 and 21: karyotype-phenotype correlations. Prenat Diagn 20: 103–122, 2000. Walters, S. (1995, 23 July). Down’s tests are a right. Sunday Age (Melbourne). Walzer S, Bashir AS, Silbert AR . Cognitive and behavioral factors in the learning disabilities of 47,XXY and 47,XYY boys. Birth Defects Orig Artic Ser 26: 45–58, 1990. Wandstrat AE, Conroy JM, Zurcher VL, et al. Molecular and cytogenetic analysis of familial Xp deletions. Am J Med Genet 94: 163–169, 2000. Wandstrat AE, Schwartz S. Isolation and molecular analysis of inv dup(15) and construction of a physical map of a common breakpoint in order to elucidate their mechanism of formation. Chromosoma 109: 498–505, 2000. Wang CM, Dixon PH, Decordova S, et al. Identification of 13 novel NLRP7 mutations in 20 families with recurrent hydatidiform mole; missense mutations cluster in the leucine-rich region. J Med Genet 46: 569–575, 2009a. Wang I, Weil D, Levilliers J, Affara NA , de la Chapelle A , Petit C. Prevalence and molecular analysis of two hot spots for ectopic recombination leading to XX maleness. Genomics 28: 52–58, 1995. Wang JC, Dang L, Fisker T. Chromosome 6 between-arm intrachromosomal insertion with intrasegmental double inversion: a four-break model. Am J Med Genet 152A : 209–211, 2010a. Wang JC, Fisker T, Dang L, Teshima I, Nowaczyk MJM. 4.3-Mb triplication of 4q32.1-q32. 2: report of a family through two generations. Am J Med Genet 149A : 2274–2279, 2009b. Wang JCC, Mamunes P, Kou SY, Schmidt J, Mao R , Hsu WT. Centromeric DNA break in a 10;16 reciprocal translocation associated with trisomy 16 confined placental mosaicism and maternal uniparental disomy for chromosome 16. Am J Med Genet 80: 418–422, 1998. Wang M, Lu S, Zhu Y, Li H. ADAM12 is an effective marker in the second trimester of pregnancy for prenatal screening of Down syndrome. Prenat Diagn 30: 561–564, 2010b. Wang MS, Schinzel A , Kotzot D, et al. Molecular and clinical correlation study of Williams-Beuren syndrome: no evidence of molecular factors in the deletion region or imprinting affecting clinical outcome. Am J Med Genet 86: 34–43, 1999a. Wang NJ, Liu D, Parokonny AS, Schanen NC. High-resolution molecular characterization of
612
•
REFERENCES
15q11-q13 rearrangements by array comparative genomic hybridization (array CGH) with detection of gene dosage. Am J Hum Genet 75: 267–281, 2004. Wang NJ, Parokonny AS, Thatcher KN, et al. Multiple forms of atypical rearrangements generating supernumerary derivative chromosome 15. BMC Genet 9: 2, 2008. Wang Q, Ratchford AM, Chi MM, et al. Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Mol Endocrinol 23: 1603–1612, 2009c. Wang W, Hu Y, Zhu H, Li J, Zhu R , Wang YP. A case of an infertile male with a small supernumerary marker chromosome negative for M-FISH and containing only heterochromatin. J Assist Reprod Genet 26: 291–295, 2009d. Wang Z, Cody JD, Leach RJ, O’Connell P. Gene expression patterns in cell lines from patients with 18q- syndrome. Hum Genet 104: 467–475, 1999b. Warburton D. Genetic factors influencing aneuploidy frequency. In: Dellarco VL, Voytek PE, Hollaender A, eds. Aneuploidy: Etiology and Mechanisms. New York: Plenum Press, 1985: 133–148. Warburton D. De novo balanced chromosome rearrangements and extra marker chromosomes identified at prenatal diagnosis: clinical significance and distribution of breakpoints. Am J Hum Genet 49: 995–1013, 1991. Warburton D. Trisomy 7 mosaicism: prognosis after prenatal diagnosis. Prenat Diagn 22: 1239–1240, 2002. Warburton D, Byrne J, Canki N. Chromosome Anomalies and Prenatal Development: An Atlas. Oxford Monographs on Medical Genetics No. 20. New York: Oxford University Press, 1991. Warburton D, Dallaire L, Thangavelu M, Ross L, Levin B, Kline J. Trisomy recurrence: a reconsideration based on North American data. Am J Hum Genet 75: 376–385, 2004. Warburton D, Kline J, Kinney A , Yu CY, Levin B, Brown S. Skewed X chromosome inactivation and trisomic spontaneous abortion: no association. Am J Hum Genet 85: 179–193, 2009. Warshawsky I, Chernova OB, Hübner CA , et al. Multiplex ligation-dependent probe amplification for rapid detection of proteolipid protein 1 gene duplications and deletions in affected males and carrier females with Pelizaeus-Merzbacher disease. Clin Chem 52: 1267–1275, 2006. Waters JJ, Campbell PL, Crocker AJM, Campbell CM. Phenotypic effects of balanced X-autosome translocations in females: a retrospective survey of 104 cases reported from UK laboratories. Hum Genet 108: 318–327, 2001.
Waters JJ, Waters KS. Trends in cytogenetic prenatal diagnosis in the UK: results from UKNEQAS external audit, 1987–1998. Prenat Diagn 19: 1023–1026, 1999. Watson AR , Taylor J, Rance CP, Bain J. Gonadal function in women treated with cyclophosphamide for childhood nephrotic syndrome: a long-term follow-up study. Fertil Steril 46: 331–333, 1986. Watson P. Legal and ethical issues in wrongful life actions. Melbourne University Law Review 26: 14, 2002. Watt AJ, Devereux FJ, Monk NA , Myers CJ, Gardner RJM. The phenotype in placental trisomy 7. Aust NZ J Obstet Gynaecol 31: 246–248, 1991. Watt JL, Ward K , Couzin DA , Stephen GS, Hill A . A paracentric inversion of 7q illustrating a possible interchromosomal effect. J Med Genet 23: 341–344, 1986. Wattanasirichaigoon D, Promsonthi P, Chuansumrit A , et al. Maternal uniparental disomy of chromosome 16 resulting in hemoglobin Bart’s hydrops fetalis. Clin Genet 74: 284–287, 2008. Webb GC, Voullaire LE, Rogers JG. Duplication of a small segment of 5p due to maternal recombination within a paracentric shift. Am J Med Genet 30: 875–881, 1988. Webb T, Clayton-Smith J, Cheng XJ, et al. Angelman syndrome with a chromosomal inversion 15 inv(p11q13) accompanied by a deletion in 15q11q13. J Med Genet 29: 921–924, 1992. Weber W. France’s highest court recognises “the right not to be born.” Lancet 358: 1972, 2001. Webley C, Halliday J. Report on Prenatal Diagnostic Testing in Victoria 2000. Melbourne, Australia: Perinatal Data Collection Unit and the Murdoch Childrens Research Institute, 2001. Wegner RD, Entezami M, Knoll U, Horn D, Sohl S, Becker R . Prenatal diagnosis of fetal trisomy 6 mosaicism and phenotype of the affected newborn. Am J Med Genet 124A : 85–88, 2004. Wegner RD, Kistner G, Becker R , et al. Fetal 46,XX/69,XXY mixoploidy: origin and confirmation by analysis of fetal urine cells. Prenat Diagn 29: 287–289, 2009. Wei HJ, Chiang HS, Lin WM, Wen JY. Pregnancy after preimplantation genetic diagnosis by fluorescence in situ hybridization using 18-, X-, and Y-chromosome probes in an infertile male with mosaic trisomy 18. J Assist Reprod Genet 17: 229–231, 2000. Weil D, Wang I, Dietrich A , Poustka A , Weissenbach J, Petit C. Highly homologous loci on the X and Y chromosomes are hot-spots for ectopic
recombinations leading to XX maleness. Nat Genet 7: 414–419, 1994. Weiss LA , Shen Y, Korn JM, et al. Association between microdeletion and microduplication at 16p11.2 and autism. N Engl J Med 358: 667–675, 2008. Weisz B, Rodeck CH. An update on antenatal screening for Down’s syndrome and specific implications for assisted reproduction pregnancies. Hum Reprod Update 12: 513–518, 2006. Weksberg R , Shuman C, Beckwith JB. BeckwithWiedemann syndrome. Eur J Hum Genet 18: 8–14, 2010. Wells D, Alfarawati S, Fragouli E. Use of comprehensive chromosomal screening for embryo assessment: microarrays and CGH. Mol Hum Reprod 14: 703–710, 2008. Wells D, Escudero T, Levy B, Hirschhorn K , Delhanty JDA , Munne S. First clinical application of comparative genomic hybridization (CGH) and polar body testing for preimplantation genetic diagnosis (PGD) of aneuploidy. Fertil Steril 78: 543–549, 2002. Wenger SL, Boone LY, Cummins JH, et al. Newborn infant with inherited ring and de novo interstitial deletion on homologous chromosome 22s. Am J Med Genet 91: 351–354, 2000. Wenger SL, Sell SL, Painter MJ, Steele MW. Inherited unbalanced subtelomeric translocation in a child with 8p- and Angelman syndromes. Am J Med Genet 70: 150–154, 1997. Wenger SL, Steele MW, Boone LY, Lenkey SG, Cummins JH, Chen XQ. “Balanced” karyotypes in six abnormal offspring of balanced reciprocal translocation normal carrier parents. Am J Med Genet 55: 47–52, 1995. Wentzel C, Fernström M, Öhrner Y, Annerén G, Thuresson AC. Clinical variability of the 22q11.2 duplication syndrome. Eur J Med Genet 51: 501–510, 2008. Weremowicz S, Sandstrom DJ, Morton CC, Niedzwiecki CA , Sandstrom MM, Bieber FR . Fluorescence in situ hybridization (FISH) for rapid detection of aneuploidy: experience in 911 prenatal cases. Prenat Diagn 21: 262–269, 2001. Wessels MW, Los FJ, Frohn-Mulder IME, Niermeijer MF, Willems PJ, Wladimiroff JW. Poor outcome in Down syndrome fetuses with cardiac anomalies or growth retardation. Am J Med Genet 116A : 147–151, 2003. West BR , Allen EF. Another previously described 21 monosomy case turns out to be an unbalanced translocation. Am J Med Genet 75: 438, 1998. Wester U, Bondeson ML, Edeby C, Annerén G. Clinical and molecular characterization of
REFERENCES
•
613
individuals with 18p deletion: a genotypephenotype correlation. Am J Med Genet 140A : 1164–1171, 2006. Westra JW, Peterson SE, Yung YC, Mutoh T, Barral S, Chun J. Aneuploid mosaicism in the developing and adult cerebellar cortex. J Comp Neurol 507: 1944–1951, 2008. Weterman MA , van Ruissen F, de Wissel M, et al. Copy number variation upstream of PMP22 in Charcot-Marie-Tooth disease. Eur J Hum Genet 18: 421–428, 2010. Wheeler M, Peakman D, Robinson A , Henry G. 45,X/46,XY mosaicism: contrast of prenatal and postnatal diagnosis. Am J Med Genet 29: 565–571, 1988. White LM, Treat K , Leff A , Styers D, Mitchell M, Knoll JHM. Exclusion of uniparental inheritance of chromosome 15 in a fetus with a familial dicentric (Y;15) translocation. Prenat Diagn 18: 111–116, 1998. Whiteford ML, Baird C, Kinmond S, Donaldson B, Davidson HR . A child with bisatellited, dicentric chromosome 15 arising from a maternal paracentric inversion of chromosome 15q. J Med Genet 37: E11, 2000. Wieczorek D, Prott EC, Robinson WP, Passarge E, Gillessen-Kaesbach G. Prenatally detected trisomy 4 and 6 mosaicism—cytogenetic results and clinical phenotype. Prenat Diagn 23: 128–133, 2003. Wieland I, Muschke P, Volleth M, et al. High incidence of familial breast cancer segregates with constitutional t(11;22)(q23;q11). Genes Chromosomes Cancer 45: 945–949, 2006. Wiersma R . True hermaphroditism in southern Africa: the clinical picture. Pediatr Surg Int 20: 363–368, 2004. Wiktor AE, Van Dyke DL. Detection of low level sex chromosome mosaicism in Ullrich-Turner syndrome patients. Am J Med Genet 138A : 259–261, 2005. Wiland E, Hobel CJ, Hill D, Kurpisz M. Successful pregnancy after preimplantation genetic diagnosis for carrier of t(2;7)(p11.2;q22) with high rates of unbalanced sperm and embryos: a case report. Prenat Diagn 28: 36–41, 2008. Wiland E, Midro AT, Panasiuk B, Kurpisz M. The analysis of meiotic segregation patterns and aneuploidy in the spermatozoa of father and son with translocation t(4;5)(p15.1;p12) and the prediction of the individual probability rate for unbalanced progeny at birth. J Androl 28: 262–272, 2007. Wilbur AK . Possible case of Rubinstein-Taybi syndrome in a prehistoric skeleton from westcentral Illinois. Am J Med Genet 91: 56–61, 2000.
614
•
REFERENCES
Wildhagen MF, van Os TAM, Polder JJ, ten Kate LP, Habbema JDF. Explorative study of costs, effects and savings of screening for female fragile X premutation and full mutation carriers in the general population. Community Genet 1: 36–47, 1998. Wiley JE, Madigan M, Christie JD, Smith AW. Dispermic chimerism with two abnormal cell lines, 47,XY, +21 and 47,XX, +12. Am J Med Genet 107: 64–66, 2002. Wilkinson TA , Crolla JA . Molecular cytogenetic characterization of three familial cases of satellited Y chromosomes. Hum Genet 91: 389–391, 1993. Willatt L, Cox J, Barber J, et al. 3q29 microdeletion syndrome: clinical and molecular characterization of a new syndrome. Am J Hum Genet 77: 154–160, 2005. Willatt L, Green AJ, Trump D. Satellites on the terminal short arm of chromosome 12 (12ps), inherited through several generations in three families: a new variant without phenotypic effect. J Med Genet 38: 723–727, 2001a. Willatt LR , Barber JCK , Clarkson A , et al. Novel deletion variants of 9q13-q21.12 and classical euchromatic variants of 9q12/qh involve deletion, duplication and triplication of large tracts of segmentally duplicated pericentromeric euchromatin. Eur J Hum Genet 15: 45–52, 2007. Willatt LR , Davison BCC, Goudie D, et al. A male with trisomy 9 mosaicism and maternal uniparental disomy for chromosome 9 in the euploid cell line. J Med Genet 29: 742–744, 1992. Willatt LR , Pearson J, Green AJ. Partial trisomy of 2p and neuroblastoma. Am J Med Genet 102: 304–305, 2001b. Willemsen M, Beunders G, Callaghan M, et al. Familial Kleefstra syndrome due to maternal somatic mosaicism for interstitial 9q34.3 microdeletions. Clin Genet 80: 31–38, 2011. Willemsen MH, Fernandez BA , Bacino CA , et al. Identification of ANKRD11 and ZNF778 as candidate genes for autism and variable cognitive impairment in the novel 16q24.3 microdeletion syndrome. Eur J Hum Genet 18: 429–435, 2010. Williams C, Sandall J, Lewando-Hundt G, Heyman B, Spencer K , Grellier R . Women as moral pioneers? Experiences of first trimester antenatal screening. Soc Sci Med 61: 1983–1992, 2005. Williams J, Dear PRF. An unbalanced t(X;10) mat translocation in a child with congenital abnormalities. J Med Genet 24: 633, 1987. Williams MS, Josephson KD, Gursoy N, JacksonCook C. Suspected gonadal mosaicism for isochromosomes 18p and 18q unsubstantiated by
fluorescence in situ hybridization analysis of sperm. Genet Med 3: 318–320, 2001. Williams NM, Zaharieva I, Martin A , et al. Rare chromosomal deletions and duplications in attention-deficit hyperactivity disorder: a genomewide analysis. Lancet 376: 1401–1408, 2010. Williamson EM, Miller JF, Seabright M. Pericentric inversion (13) with two different recombinants in the same family. J Med Genet 17: 309–312, 1980. Willis MJ, Bird LM, Dell’Aquilla M, Jones MC. Expanding the phenotype of mosaic trisomy 20. Am J Med Genet 146: 330–336, 2008. Willis MJH, Bird LM, Dell’Aquilla M, Jones MC. Natural history of prenatally diagnosed 46,X, isodicentric Y. Prenat Diagn 26: 134–137, 2006. Wilson LC, Leverton K , Oude Luttikhuis MEM, et al. Brachydactyly and mental retardation: an Albright hereditary osteodystrophy-like syndrome localized to 2q37. Am J Hum Genet 56: 400–407, 1995. Wilson M, Peters G, Bennetts B, et al. The clinical phenotype of mosaicism for genome-wide paternal uniparental disomy: two new reports. Am J Med Genet 146A : 137–148, 2008. Wilson MG, Lin MS, Fujimoto A , Herbert W, Kaplan FM. Chromosome mosaicism in 6,000 amniocenteses. Am J Med Genet 32: 506–513, 1989. Wilson ND, Ross LJN, Close J, Mott R , Crow TJ, Volpi EV. Replication profile of PCDH11X and PCDH11Y, a gene pair located in the nonpseudoautosomal homologous region Xq21.3/ Yp11.2. Chromosome Res 15: 485–498, 2007. Wilton L. Preimplantation genetic diagnosis for aneuploidy screening in early human embryos: a review. Prenat Diagn 22: 512–518, 2002. Wilton L, Thornhill A , Traeger-Synodinos J, Sermon KD, Harper JC. The causes of misdiagnosis and adverse outcomes in PGD. Hum Reprod 24: 1221–1228, 2009. Wilton L, Williamson R , McBain J, Edgar D, Voullaire L. Birth of a healthy infant after preimplantation confirmation of euploidy by comparative genomic hybridization. N Engl J Med 345: 1537–1541, 2001. Wimalasundera RC, Gardiner HM. Congenital heart disease and aneuploidy. Prenat Diagn 24: 1116–1122, 2004. Wimplinger I, Morleo M, Rosenberger G, et al. Mutations of the mitochondrial holocytochrome c-type synthase in X-linked dominant microphthalmia with linear skin defects syndrome. Am J Hum Genet 79: 878–889, 2006. Winter JSD. Androgen therapy in Klinefelter syndrome during adolescence. Birth Defects Orig Artic Ser 26: 235–245, 1990.
Winther JF, Boice JD, Jr., Mulvihill JJ, et al. Chromosomal abnormalities among offspring of childhood-cancer survivors in Denmark: a population-based study. Am J Hum Genet 74: 1282–1285, 2004. Witters I, Devriendt K , Legius E, et al. Rapid prenatal diagnosis of trisomy 21 in 5049 consecutive uncultured amniotic fluid samples by fluorescence in situ hybridisation (FISH). Prenat Diagn 22: 29–33, 2002. Witters I, Fryns JP. Follow-up of a child with trisomy 17 mosaicism. Prenat Diagn 28: 1080, 2008. Wolff DJ, Miller AP, Van Dyke DL, Schwartz S, Willard HF. Molecular definition of breakpoints associated with human Xq isochromosomes: implications for mechanisms of formation. Am J Hum Genet 58: 154–160, 1996. Wolstenholme J. Confined placental mosaicism for trisomies 2, 3, 7, 8, 9, 16, and 22: their incidence, likely origins, and mechanisms for cell lineage compartmentalization. Prenat Diagn 16: 511–524, 1996. Wolstenholme J, Angell RR . Maternal age and trisomy—a unifying mechanism of formation. Chromosoma 109: 435–438, 2000. Wolstenholme J, Brummitt JA , English CJ, Goodship JA . Prenatal detection of multiple copies of a familial supernumerary marker chromosome. Prenat Diagn 12: 1067–1071, 1992. Wolstenholme J, Emslie JB, Connors S. Association of Clinical Cytogeneticists chorion villus sampling database 1987–2000. Prenat Diagn 26: 420–427, 2006. Wolstenholme J, Webb AL, English CJ, Evans J. Prenatal diagnosis of chromosome 22 mosaicism with extensive investigation of placental and fetal tissues. Ann Génét 44(suppl 1): 14s, 2001a. Wolstenholme J, White I, Sturgiss S, Carter J, Plant N, Goodship JA . Maternal uniparental heterodisomy for chromosome 2: detection through “atypical” maternal AFP/hCG levels, with an update on a previous case. Prenat Diagn 21: 813–817, 2001b. Won RH, Currier RJ, Lorey F, Towner DR . The timing of demise in fetuses with trisomy 21 and trisomy 18. Prenat Diagn 25: 608–611, 2005. Wong EC, Ferguson KA , Chow V, Ma S. Sperm aneuploidy and meiotic sex chromosome configurations in an infertile XYY male. Hum Reprod 23: 374–378, 2008. Woodage T, Prasad M, Dixon JW, et al. Bloom syndrome and maternal uniparental disomy for chromosome 15. Am J Hum Genet 55: 74–80, 1994.
REFERENCES
•
615
Woods CG, Bankier A , Curry J, et al. Asymmetry and skin pigmentary anomalies in chromosome mosaicism. J Med Genet 31: 694–701, 1994. Woods CG, Noble J, Falconer AR . A study of brothers with Klinefelter syndrome. J Med Genet 34: 702, 1997. Woods KS, Cundall M, Turton J, et al. Over- and underdosage of SOX3 is associated with infundibular hypoplasia and hypopituitarism. Am J Hum Genet 76: 833–849, 2005. Woodward K , Kendall E, Vetrie D, Malcolm S. Pelizaeus-Merzbacher disease: identification of Xq22 proteolipid-protein duplications and characterization of breakpoints by interphase FISH. Am J Hum Genet 63: 207–217, 1998. Worley KC, Lindsay EA , Bailey W, Wise J, McCabe ERB, Baldini A . Rapid molecular cytogenetic analysis of X-chromosomal microdeletions: fluorescence in situ hybridization (FISH) for complex glycerol kinase deficiency. Am J Med Genet 57: 615–619, 1995. Worsham MJ, Miller DA , Devries JM, et al. A dicentric recombinant 9 derived from a paracentric inversion: phenotype, cytogenetics, and molecular analysis of centromeres. Am J Hum Genet 44: 115–123, 1989. Worton RG, Stern R . A Canadian collaborative study of mosaicism in amniotic fluid cell cultures. Prenat Diagn 4(spec no.): 131–144, 1984. Wray AM, Dennis TR , Ghidini A , Gorman B, Haddad BR , Meck JM. Prenatal diagnosis of supernumerary ring chromosome 1: case report and review of the literature. Genet Couns 18: 233–241, 2007. Wright CF, Chitty LS. Cell-free fetal DNA and RNA in maternal blood: implications for safer antenatal testing. Br Med J 339: b2451, 2009. Wu DJ, Wang NJ, Driscoll J, et al. Autistic disorder associated with a paternally derived unbalanced translocation leading to duplication of chromosome 15pter-q13. 2: a case report. Mol Cytogenet 2: 27, 2009. Wu YC, Yu MT, Chen LC, Chen CL, Yang ML. Prenatal diagnosis of mosaic tetrasomy 10p associated with megacisterna magna, echogenic focus of left ventricle, umbilical cord cysts and distal arthrogryposis. Am J Med Genet 117A : 278–281, 2003. Wu YQ, Badano JL, McCaskill C, Vogel H, Potocki L, Shaffer LG. Haploinsufficiency of ALX4 as a potential cause of parietal foramina in the 11p11.2 contiguous gene-deletion syndrome. Am J Hum Genet 67: 1327–1332, 2000. Wu YQ, Nickerson E, Shaffer LG, Keppler-Noreuil K , Muilenburg A . A case of Williams syndrome with
616
•
REFERENCES
a large, visible cytogenetic deletion. J Med Genet 36: 928–932, 1999. Wyrobek AJ, Adler ID. Detection of aneuploidy in human and rodent sperm using FISH and applications of sperm assays of genetic damage in heritable risk evaluation. Mutat Res 352: 173–179, 1996. Xu J, Chernos J, Roland B. Trisomy 16pter to 16q12.1 and monosomy 22pter to 22q11.2 resulting from adjacent-2 segregation of a maternal complex chromosome rearrangement. Am J Med Genet 73: 327–329, 1997. Xu S, Han JC, Morales A , Menzie CM, Williams K , Fan YS. Characterization of 11p14-p12 deletion in WAGR syndrome by array CGH for identifying genes contributing to mental retardation and autism. Cytogenet Genome Res 122: 181–187, 2008. Xu W, Robert C, Thornton PS, Spinner NB. Complete androgen insensitivity syndrome due to X chromosome inversion: a clinical report. Am J Med Genet 120A : 434–436, 2003. Yaegashi N, Senoo M, Uehara S, et al. Age-specific incidences of chromosome abnormalities at the second trimester amniocentesis for Japanese mothers aged 35 and older: collaborative study of 5484 cases. J Hum Genet 43: 85–90, 1998. Yamanaka M, Setoyama T, Igarashi Y, et al. Pregnancy outcome of fetuses with trisomy 18 identified by prenatal sonography and chromosomal analysis in a perinatal center. Am J Med Genet 140: 1177–1182, 2006. Yan D, Ouyang XM, Angeli SI, Du LL, Liu XZ. Paternal uniparental disomy of chromosome 13 causing homozygous 35delG mutation of the GJB2 gene and hearing loss. Am J Med Genet 143A : 385–386, 2007. Yang Q, Sherman SL, Hassold TJ, et al. Risk factors for trisomy 21: maternal cigarette smoking and oral contraceptive use in a population-based casecontrol study. Genet Med 1: 80–88, 1999. Yang SP, Bidichandani SI, Figuera LE, et al. Molecular analysis of deletion (17)(p11.2p11.2) in a family segregating a 17p paracentric inversion: implications for carriers of paracentric inversions. Am J Hum Genet 60: 1184–1193, 1997. Yardin C, Esclaire F, Laroche C, et al. Should the chromosome region 15q11q13 be tested systematically by FISH in the case of an autisticlike syndrome? Clin Genet 61: 310–313, 2002. Yardin C, Esclaire F, Terro F, Baclet MC, Barthe D, Laroche C. First familial case of ring chromosome 18 and monosomy 18 mosaicism. Am J Med Genet 104: 257–259, 2001. Yaron Y, Carmon E, Goldstein M, et al. The clinical application of spectral karyotyping (SKY) in the
analysis of prenatally diagnosed extra structurally abnormal chromosomes (ESACs). Prenat Diagn 23: 74–79, 2003. Yaron Y, Feldman B, Kramer RL, et al. Prenatal diagnosis of 46,XY/46,XX mosaicism: a case report. Am J Med Genet 84: 12–14, 1999. Yenamandra A , Deangelo P, Aviv H, Suslak L, Desposito F. Interstitial insertion of Y-specific DNA sequences including SRY into chromosome 4 in a 45,X male child. Am J Med Genet 72: 125–128, 1997. Yeung A , Bruno D, Scheffer IE, et al. 4.45 Mb microduplication in chromosome band 14q12 including FOXG1 in a girl with refractory epilepsy and intellectual impairment. Eur J Med Genet 52: 440–442, 2009a. Yeung A , Francis D, Giouzeppos O, Amor DJ. Pallister-Killian syndrome caused by mosaicism for a supernumerary ring chromosome 12p. Am J Med Genet 149A : 505–509, 2009b. Yip MY, Messias A , Dalzell P, Kovacic A , Saville T, Purvis-Smith S. Unbalanced translocation mosaicism in a woman with recurrent miscarriages. Bull Hum Genet Soc Australasia 9: 44, 1996. Yobb TM, Somerville MJ, Willatt L, et al. Microduplication and triplication of 22q11. 2: a highly variable syndrome. Am J Hum Genet 76: 865–876, 2005. Yokoyama Y, Narahara K , Teraoka M, et al. Cryptic pericentric inversion of chromosome 17 detected by fluorescence in situ hybridization study in familial Miller-Dieker syndrome. Am J Med Genet 71: 236–237, 1997. Yong PJ, Barrett IJ, Kalousek DK , Robinson WP. Clinical aspects, prenatal diagnosis, and pathogenesis of trisomy 16 mosaicism. J Med Genet 40: 175–182, 2003. Yong PJ, Langlois S, von Dadelszen P, Robinson W. The association between preeclampsia and placental trisomy 16 mosaicism. Prenat Diagn 26: 956–961, 2006. Yong PJ, Marion SA , Barrett IJ, Kalousek DK , Robinson WP. Evidence for imprinting on chromosome 16: the effect of uniparental disomy on the outcome of mosaic trisomy 16 pregnancies. Am J Med Genet 112: 123–132, 2002. Yoon PW, Freeman SB, Sherman SL, et al. Advanced maternal age and the risk of Down syndrome characterized by the meiotic stage of chromosomal error: a population-based study. Am J Hum Genet 58: 628–633, 1996. Youings S, Ellis K , Ennis S, Barber J, Jacobs P. A study of reciprocal translocations and inversions detected by light microscopy with special reference
to origin, segregation, and recurrent abnormalities. Am J Med Genet 126A : 46–60, 2004. Yu S, Cox K , Friend K , et al. Familial 22q11.2 duplication: a three-generation family with a 3-Mb duplication and a familial 1.5-Mb duplication. Clin Genet 73: 160–164, 2008. Yue Y, Farcas R , Thiel G, et al. De novo t(12;17) (p13.3;q21.3) translocation with a breakpoint near the 5’ end of the HOXB gene cluster in a patient with developmental delay and skeletal malformations. Eur J Hum Genet 15: 570–577, 2007. Zackai EH, Emanuel BS. Site-specific reciprocal translocation, t(11;22) (q23;q11), in several unrelated families with 3:1 meiotic disjunction. Am J Med Genet 7: 507–521, 1980. Zahed L, Der Kaloustian V, Batanian JR . Familial complex chromosome rearrangement giving rise to balanced and unbalanced recombination products. Am J Med Genet 79: 30–34, 1998. Zaragoza MV, Keep D, Genest DR , Hassold T, Redline RW. Early complete hydatidiform moles contain inner cell mass derivatives. Am J Med Genet 70: 273–277, 1997. Zaragoza MV, Surti U, Redline RW, Millie E, Chakravarti A , Hassold TJ. Parental origin and phenotype of triploidy in spontaneous abortions: predominance of diandry and association with the partial hydatidiform mole. Am J Hum Genet 66: 1807–1820, 2000. Zaslav AL, Blumenthal D, Willner JP, Pierno G, Jacob J, Fox JE. Prenatal diagnosis of trisomy 4 mosaicism. Am J Med Genet 95: 381–384, 2000. Zaslav AL, Fallet S, Blumenthal D, Jacob J, Fox J. Mosaicism with a normal cell line and an unbalanced structural rearrangement. Am J Med Genet 82: 15–19, 1999. Zaslav AL, Fallet S, Brown S, et al. Prenatal diagnosis of low level trisomy 15 mosaicism: review of the literature. Clin Genet 53: 286–292, 1998. Zaslav AL, Fox JE, Jacob J, et al. Significance of a prenatally diagnosed del(10)(q23). Am J Med Genet 107: 174–176, 2002. Zaslav AL, Pierno G, Davis J, et al. Prenatal diagnosis of trisomy 3 mosaicism. Prenat Diagn 24: 693–696, 2004. Zatsepin I, Verger P, Robert-Gnansia E, et al. Down syndrome time-clustering in January 1987 in Belarus: link with the Chernobyl accident? Reprod Toxicol 24: 289–295, 2007. Zayed F, Ghalayini I, Matalka I. A male phenotype (XY) hermaphrodite treated for seminoma, fathered a healthy child by IVF-ICSI technique. J Assist Reprod Genet 25: 345–348, 2008.
REFERENCES
•
617
Zech NH, Wisser J, Natalucci G, Riegel M, Baumer A , Schinzel A . Monochorionic-diamniotic twins discordant in gender from a naturally conceived pregnancy through postzygotic sex chromosome loss in a 47,XXY zygote. Prenat Diagn 28: 759–763, 2008. Zechner U, Kohlschmidt N, Rittner G, et al. Epimutation at human chromosome 14q32.2 in a boy with a upd(14)mat-like clinical phenotype. Clin Genet 75: 251–258, 2009. Zeesman S, Zwaigenbaum L, Whelan DT, Hagerman RJ, Tassone F, Taylor SA . Paternal transmission of fragile X syndrome. Am J Med Genet 129A : 184–189, 2004. Zellweger H, Abbo G. Familial mosaicism attributable to a new gene. Lancet 1: 455–457, 1965. Zeng S, Patil SR , Yankowitz J. Prenatal detection of mosaic trisomy 1q due to an unbalanced translocation in one fetus of a twin pregnancy following in vitro fertilization: a postzygotic error. Am J Med Genet 120A : 464–469, 2003. Zenker M, Wermuth B, Trautmann U, et al. Severe, neonatal-onset OTC deficiency in twin sisters with a de novo balanced reciprocal translocation t(X;5) (p21.1;q11). Am J Med Genet 132A : 185–188, 2005. Zeschnigk M, Albrecht B, Buiting K , et al. IGF2/H19 hypomethylation in Silver-Russell syndrome and isolated hemihypoplasia. Eur J Hum Genet 16: 328–334, 2008. Zhang A , Weaver DD, Palmer CG. Molecular cytogenetic identification of four X chromosome duplications. Am J Med Genet 68: 29–38, 1997. Zhang F, Khajavi M, Connolly AM, Towne CF, Batish SD, Lupski JR . The DNA replication FoSTeS/ MMBIR mechanism can generate genomic, genic and exonic complex rearrangements in humans. Nat Genet 41: 849–853, 2009a. Zhang L, Coffey S, Lua LL, et al. FMR1 premutation in females diagnosed with multiple sclerosis. J Neurol Neurosurg Psychiatry 80: 812–814, 2009b. Zhang YX , Zhang YP, Gu Y, et al. Genetic analysis of first-trimester miscarriages with a combination of cytogenetic karyotyping, microsatellite genotyping and arrayCGH. Clin Genet 75: 133–140, 2009c.
618
•
REFERENCES
Zinn AR , Ouyang B, Ross JL, Varma S, Bourgeois M, Tonk V. Del (X)(p21.2) in a mother and two daughters with variable ovarian function. Clin Genet 52: 235–239, 1997. Zollino M, Lecce R , Fischetto R , et al. Mapping the Wolf-Hirschhorn syndrome phenotype outside the currently accepted WHS critical region and defining a new critical region, WHSCR-2. Am J Hum Genet 72: 590–597, 2003. Zollino M, Murdolo M, Marangi G, et al. On the nosology and pathogenesis of Wolf-Hirschhorn syndrome: genotype-phenotype correlation analysis of 80 patients and literature review. Am J Med Genet 148C: 257–269, 2008. Zollino M, Orteschi D, Marangi G, et al. A case of Beckwith-Wiedemann syndrome caused by a cryptic 11p15 deletion encompassing the centromeric imprinted domain of the BWS locus. J Med Genet 47: 429–432, 2010a. Zollino M, Orteschi D, Neri G. Phenotypic map in ring 14 syndrome. Am J Med Genet 152A : 237, 2010b. Zori RT, Boyar FZ, Williams WN, et al. Prevalence of 22q11 region deletions in patients with velopharyngeal insufficiency. Am J Med Genet 77: 8–11, 1998. Zou YS, Newton S, Milunsky JM. A complex maternal rearrangement results in a pure 10.8 Mb duplication of the 5q13.1-q14.1 region in an affected son. Am J Med Genet 152A : 498–503, 2010. Zou YS, Van Dyke DL, Thorland EC, et al. Mosaic ring 20 with no detectable deletion by FISH analysis: characteristic seizure disorder and literature review. Am J Med Genet 140: 1696–1706, 2006. Zuccarello D, Dallapiccola B, Novelli A , Foresta C. Azoospermia in a man with a constitutional ring 22 chromosome. Eur J Med Genet 53: 389–391, 2010. Zuffardi O, Bonaglia M, Ciccone R , Giorda R . Inverted duplications deletions: underdiagnosed rearrangements? Clin Genet 75: 505–513, 2009. Zühlke C, Thies U, Braulke I, Reis A , Schirren C. Down syndrome and male fertility: PCR-derived fingerprinting, serological and andrological investigations. Clin Genet 46: 324–326, 1994.
INDEX
1qh variation, 258 3:1 segregation autosomal reciprocal translocation, 71, 86, 87, 89, 107 X-autosome translocation, 128 4:0 segregation autosomal reciprocal translocation, 71, 89 X-autosome translocation, 128 4;8 translocation, 80 11;22 translocation, 86 breast cancer, 86, 107 embryos, 437 risks to carrier, 107 22q11 deletion syndrome, 326 45,X See Turner syndrome 45,X male, 336 Y-autosome translocation, 132 Xp-Yp translocation, 134 45,X/46,XX mosaicism in normal women, 225 45,X/46,XY mosaicism, 228 45,X/47,XXY mosaicism, 228 46,XX male See XX male 48,XXXX See Polysomy sex chromosomes 48,XXXY See Polysomy sex chromosomes 48,XXYY See Polysomy sex chromosomes 48,XYYY See Polysomy sex chromosomes 49,XXXXX See Polysomy sex chromosomes 49,XXXXY See Polysomy sex chromosomes 49,XXXYY See Polysomy sex chromosomes 49,XXYYY See Polysomy sex chromosomes 49,XYYYY See Polysomy sex chromosomes 68,XX See hypotriploidy 69,XXX, 69,XXY See triploidy
Abbreviations, 505 Abortion genetic, 16 attitudes, 16 decision-making , 439 mental retardation, 16 in the mentally retarded, 17 occult, 382 recurrent See Recurrent miscarriage spontaneous See Miscarriage Acardiac monster, 386 Acrocentric chromosomes short arm variation, 259 significance, 260 Adjacent segregation, rob, 144 Adjacent-1 segregation autosomal, 71, 78, 80, 81, 102 X-autosomal, 124 Adjacent-2 segregation autosomal, 82, 106 X-autosomal, 124 Agent Orange, 496 Aging and chromosome loss, 32 Alagille syndrome, 325 Albright-like syndrome with deletion 2q, 312 Alcohol, risks to offspring , 496 Alpha thalassemia-retardation, del(16p), 321 Alternate segregation reciprocal translocation, 71 rob translocation, 144 Alzheimer’s disease, association with DS, 280 Amniocentesis See Prenatal diagnosis
•
619
Analphoid centromere See Neocentromere Anaphase lag , 42 Androgen insensitivity syndrome, 335 genetic counseling , 339 Aneusomie de recombinaison, 164 Angell’s hypothesis, 37 Angelman syndrome, 362 critical region, 56 deletion, 362 due to rearrangement, 363 imprinting center defects, 362, 366 interchange trisomy, rescue from IVF association, 372 microdeletion, 372 non-deletion, non-UPD, 366 risks to parents of AS child, 364, 371 simplification of genetics, 373 UBE3A mutation, 366, 372 UPD, 364 Antimicrobial therapy, 494 Apparently balanced rearrangement, 45, 95, 308 but actually unbalanced, 308 de novo, 95 locus disruption, 308 prenatal diagnosis, 466 unmasking recessive gene, 300 Array, See Microarray Ascertainment bias, 60 Association, coincidental or causal, 62 Ataxia-telangiectasia, 344 cancer risk in heterozygote, 345 Autism microdeletion 16p11.2, 273 microduplication 16p11.2, 238 AZF loci, 391 Azoöspermia, definitions, 391 Banking gonadal tissue, 493 Barr body, 222 Beckwith-Wiedemann syndrome, 360 epigenetic error, 360, 369 IVF association, 369 mendelian mutation, 369 risks to parent of BWS child, 369 segmental UPD, 360 Benzodiazepine treatment, 494 Biallelic expression, 353 Bisphenol A , 496 Blaschko, lines of, 53 Blastocyst biopsy, 430 karyotyping , 381 Blastomere biopsy, 430 Blighted ovum, 382 Bloom syndrome, 343 prenatal diagnosis, 344 Breakpoints disrupting loci See Locus disruption Breast cancer risk ataxia-telangiectasia carrier, 345 t(11q;22q) carrier, 86 BRCA2, Fanconi anemia, 342 Buccal smear, 222 Café-au-lait macules, ring chromosome, 202 Caffeine, risks to offspring , 496 Campomelic dysplasia, 308, 338 Cancer androgen insensitivity, risk in, 335
620
•
INDEX
ataxia-telangiectasia carrier, risks to, 345 breast, with t(11;22), 86 chemotherapy See Cancer therapy childhood survivors, 490 deletion 5q and adenomatous polyposis, 315 deletion 9q and Gorlin syndrome, 315, 318 deletion 10q and juvenile polyposis, 315 deletion 13q and retinoblastoma, 319 deletion 17p13 and Li-Fraumeni syndrome, 16 gonadal tissue banking , 494 hydatidiform mole, 400 kidney, chromosome 3 translocation, 111 reciprocal translocation, 111 sperm banking , 494 therapy, risks to offspring chemotherapy, 490 hydatidiform mole, 493 leukemia, 493 lymphoma, 492 radioisotopes, 492 radiotherapy, 492 seminoma, 493 testicular cancer, 493 Wilms tumor, 493 trisomy 8 mosaicism, 241 t(1;17) and neuroblastoma, 111 XY female, 339 Cat-eye syndrome See Isodicentric 22 C-banding , 21 significance, 259 variation, 257 Y chromosome, 259 CCG repeat See Trinucleotide repeat expansion Celocentesis, 424 Centric fission, 155 risks to carrier, 159 rob, 159 Centromere, 6 neocentromere, 307 premature centromere separation, 345, 346 Centromere fission, 155, 159 Centromere-telomere fusion, 308 Cervical lavage, 424 CGH See Comparative genomic hybridization CGH-array, See Microarray Chaotic mosaicism of embryo, 33, 428, 435 Charcot-Marie-Tooth neuropathy, 330 CHARGE association, 317 Chemotherapy See Cancer Chernobyl accident, 495 Children, testing of, 14 Chimerism, 33 ovotesticular disorder sex development, 337 Chorionic villus sampling See Prenatal diagnosis Choroid plexus cysts, 417 Chromosome breakage syndromes, 341 ovarian failure, 347 Chromosome polymorphisms, 257 CNV See Copy number variant Coffee drinking See Caffeine Coffin-Siris syndrome, with translocation, 97 Comparative genomic hybridization, 22, 24 Complementary isochromosome, 157, 159 Complementary deletion/duplication, 298 Complementation, gametic, 37 cause of UPD, 353 Complete hydatidiform mole See Hydatidiform mole Complex chromosome rearrangement, 212
categories, 212 cryptic, 216 de novo, prenatal diagnosis, 468 double two-way, 212, 217 exceptional CCR , 212, 215 infertility, 219 ISCN nomenclature, 212 preimplantation diagnosis, 220 prenatal diagnosis, 220 risks to carrier, 219 segregation, 213 three-way, 212, 213 Confidence limits, 510 Confined placental mosaicism, 56, 442, 449, 456 placental effects, 452 Congenital insensitivity to pain, isozygosity, 352 Constitutional abnormality, 7 Contiguous gene syndrome, 299 Copy number change, 270 Copy number variants, 269 definitions, 270 normal variants, 271 variable penetrance of, 63 Cordocentesis See Fetal blood sampling Cornelia de Lange syndrome, 345 Correction of monosomy, 44 rob, 148, 149 Correction of trisomy, 42, 353 prenatal diagnosis, 444 rob, 145 Couple both with translocation, 97, 149 Countertype phenotypes, 47 Cri du chat syndrome, 313 Critical regions X chromosome See X critical regions Cryptic rearrangement, 109 Cure, triploidy, 293 Cutis tricolor, 53 CVS See Chorionic villus sampling Cystic hygroma, 424 Database resources, 51 DAZ (deleted in azoöspermia) gene, 391 DECIPHER , 274, 309 Delayed digyny, diploid/triploid mosaicism, 289 Delayed dispermy, diploid/triploid mosaicism, 289 Deletion, 299, 308 de novo origin, 308 generation, 296 familial transmission, 235 microarray detection, 309 recurrence risk, 308 risks to carrier, 235 subtelomeric, 300 unmasking recessive gene, 300 without phenotypic effect, 265 Deletions of individual chromosomes: 1p36.3, 311 1q21.1, 311 TAR syndrome, 273 1q41q42, 312 2q23.1, 312 2q37, Albright-like, 312 3p26.2, 312 3q29, 312 4p, Wolf-Hirschhorn syndrome, 312 4q34, 313 5p, cri du chat syndrome, 313 5p13.1, Cornelia de Lange syndrome, 313
5q22q23, polyposis plus, 315 5q35, Sotos syndrome, 315 6q, 315 7q11.23, Williams syndrome, 316 7p13, Greig syndrome, 316 7p21.1, Saethre-Chotzen syndrome, 316 7q21.3, ectrodactyly plus, 316 7q32, holoprosencephaly plus, 317 8p23.1, 317 normal variant, 264 8q12, CHARGE, 317 8q24, Langer-Giedion syndrome, 317 9p22p23, 317 9p24.3, sex reversal, 317 9q22q31, 318 9q34.3, Kleefstra syndrome, 318 10p13, DiGeorge phenocopy, 318 10q11.2, Hirschsprung plus, 318 11p11.2, Potocki-Shaffer syndrome, 318 11p13, WAGR syndrome, 318 11q23, Jacobsen syndrome, 319 13q14, retinoblastoma plus, 319 13q21.1, 301, 319 14q12, ‘congenital Rett syndrome’, 319 15q11.2 See Prader-Willi/Angelman syndromes 15q13.2-13.3, 319 specific treatment, 320 16p11.2, 273, 321 16p12.1, 321 16p13.11, 321 16p13.3, ATR-16, 321 16p13.3, Rubinstein-Taybi syndrome, 321 16q24 syndromes, 322 17p11.2, Smith-Magenis syndrome, 323 17p12, pressure-sensitive neuropathy, 323 17p13.1, 323 17p13.3, Miller-Dieker syndrome, 322 17q21.31, 325 17q23, 325 18p, 18q, 325 19p13, 325 19q13.11, 325 20p12, Alagille syndrome, 325 20q13.33, 326 21q, 326 22q11, DiGeorge syndrome, 326 22q11.2, 327 22q13.3, Phelan-McDermid syndrome, 327 X , 226, 231 microdeletion, 228, 232 Xp22.3, ichthyosis, 328 Xp22.3, Kallmann syndrome, 328 De novo apparently balanced rearrangement See Apparently balanced rearrangement Developmental delay, 9 Diabetes risk to child, 493 transient neonatal, 359 Diagnostic X-rays, risk to child, 494 Diandry, 287, 394 Diazepam, sperm studies, 494 ‘Dieggy’, 288 DiGeorge syndrome, 326 Digynic triploidy, 288 Diploid/tetraploid mosaicism, 289 Diploid/triploid mosaicism, 289 Disorders of sex development, 333 extragonadal defects, 338
INDEX
•
621
Disomy X See X disomy Dispermy, 287, 395 Disruption of loci at breakpoint, 308 Dissatisfied life, 18 Distamycin A/DAPI staining , 22 Donor oöcyte See Ovum donor Dosage effect, 7, 278 Double aneuploidy, 36, 63 Double segment exchange autosomal translocation, 68, 80, 84 subtelomeric, 81 X-autosome translocation, 124 Double two-way translocation, 212, 217 Down syndrome See Trisomy 21 Duplication, 302 familial transmission, 237 microarray, 238 mosaicism, 239 risks to carrier, 237 tandem, 303 without phenotypic effect, 265 Duplications of individual chromosomes: 1q21.1, 328 2p22p25, 328 2q37, 328 3q29, 328 7q11.23, 329 8p23.1, normal variant, 265 10q24, split hand/foot, 329 15q11q13.1, 329 15q11.2, normal variant, 238, 264 15q13.2q13.3, 330 15q26, 330 16p11.2, 238, 330 normal variants, 265 16p13.11, 330 16p13.3, 330 17p11.2, Potocki-Lupski syndrome, 331 17p12, Charcot-Marie-Tooth, 330 17p13.3, 330 17q21.31, 331 22q11.2, 331 22q13, 331 X , 229, 232 Xp11.22-p11.23, 229, 331 Xq22, Pelizaeus-Merzbacher, 331 Xq27.1, panhypopituitarism, 332 Xq28, Lubs syndrome, 332 Duplication/deletion double segment translocation, 80 Duplicon, 296 Dynamic mosaicism jumping translocation, 159 ring , 202 Early amniocentesis, 423 ECARURA , 274, 309, 310 Eclampsia, trisomy 16 mosaicism, 463 Ectrodactyly, 316 Edwards syndrome See Trisomy 18 Egg See Oöcyte Emanuel syndrome, 86, 100 Embryo chaotic mosaicism, 435 development, 442 fetus papyraceous, 385 karyotyping , 380, 382 monosomy, 381
622
•
INDEX
moral status, 18 mosaicism, 381 phenotypes, abnormal, 385 translocation carriers (Table), 76, 437 trisomy 8 phenotype, 382 Embryology, 442 Empiric risks, 59 Empty sac, 382 Environmental chromosome damage, 489 Epidemiology of trisomy 21, 407 Epigenetics, 45, 353 IVF, 394 Epilepsy chromosomal, 48 ring 20, 206 Epimutation, 357 Angelman, 365, 372, 394 Beckwith-Wiedemann, 369, 394 Prader-Willi, 365, 371 Silver-Russell, 369 Euchromatin, 6 Euchromatic variants, 263 advice, 267 4p16.1, 264 8p23.1, 264 pericentromeric 9, 264 15q11.2, 264 16p11.2, 265 16q11.2, 265 subtelomeric, 266 EUCROMIC CVS data, 451 Exostoses, in deletion syndromes, 317, 318 Expansion and See Trinucleotide repeat expansion duplication to triplication, 267 Extra structurally abnormal chromosome See Supernumerary marker chromosome Family, ‘duty’ to be tested, 13 Fanconi anemia, 342 prenatal diagnosis, 343 Fetal blood sampling , 423 Fetal cells from maternal blood, 423 Fetal death in utero, 386, 397 Fetal nasal bone, 411 Fetal ultrasonography, 411, 417 Fetus papyraceus, 385 FISH, 22 Fission See Centric fission Fluorescence in situ hybridization, 22 Folic acid, 494 Fork stalling and template switching , 297 Fragile sites, 260 clinical significance, 261, 268 Fragile XA , 242 brain pathology, 245 carrier frequency, 243 compound heterozygote, 250 cytogenetics, 250 expansion of mutation, 244, 250 FMR1 gene, 242 deletion, 249 expansion sizes, 243 frequencies, 243 methylation, 245 FMRP protein, 242 frequency, 243 full mutation, 243 gray zone allele, 243, 248, 253
intermediate allele, 253 expansion, 243, 248 laboratory detection, 245 mosaicism methylation, 249 mutational, 249 population screening , 215 predictive gene testing , 15 preimplantation diagnosis, 253 premature ovarian failure, 248 premutation, 243 ‘high-end’, ‘low-end’, 253 ovarian failure, 248 RNA toxicity, 246 prenatal diagnosis, 254 risks to carrier, 250, 252 screening , 215 treatment, 254 tremor-ataxia syndrome, 247, 248 Fragile XD, 242 Fragile XE, 242 frequency, 243 molecular genetics, 249 prenatal diagnosis, 254 risks to carrier, 253 segregation, 250 Fragile XF, 242, 249 Frequencies chromosomal abnormalities, 8, 11, 12, 13 Functional X disomy X-autosome translocation, 115, 117, 119 Gamete donation See Ovum donor, Sperm donor Gametic complementation See Complementation, gametic Gautier Marie, 3 G-banding , 21 Gene-rich and sparse regions, 47 General ring syndrome, 202 Genetic abortion, 16 Genitopatellar syndrome, 338 Genoglyphics, 274, 310 Genomic disorders, 296 Genomic imprinting See Imprinting Genotype-first approach, 11 Giemsa banding , 21 Gonadal dysgenesis pure XY, 334 rare forms, 338 Gonadal mosaicism, 54 Hultén’s hypothesis, 54 isochromosomes, 305 ovarian karyotyping , 54, 282 rob isochromosome, 283 sperm karyotyping , 54, 306 SRY gene, 334 translocation, 239 trisomy 21, 54, 282 Greig syndrome, 316 Grey zone fragile X allele, 243, 248, 253 Griseofulvin, 494 ‘Guilt’ in carriers, 16 Habitual abortion See Recurrent miscarriage Haploid autosomal length, 499 Haplo-insufficiency, 299 HC Forum, 101 HDR syndrome, 318 Heart defects, 48
Hereditary neuropathy with pressure palsies, 323 Hermaphroditism See Ovotesticular disorder of sex development Heterochromatin, 6, 258 instability, in ICF syndrome, 346 repulsion, in Roberts syndrome, 345 translocation of Yqh, 261 Heterologous rob See Robertsonian translocation, heterologous Heteromorphisms, 258 Heterosynapsis, 30 insertion, 184 inversion, 165 translocation, 94 Heterotrisomy, 56 Heterozygote, definition, 5 Hexasomy, 307 Hiroshima bombing , 495 Hirschsprung plus syndrome, 318 History of ‘chromosome’, 3 Hodgkin’s disease, 492 Homologous rob See Robertsonian translocation, homologous Homosynapsis, 30 in inversion, 164 Hultén hypothesis in re trisomy 21, ‘Humster’ sperm analysis technique, 379 Hydatidiform mole, 394 biparental inheritance, 395, 400 cancer risk, 400 complete, 394 frequency, 395 partial, 395 recurrence, 400 uniparental origin, 394 Hypomelanosis of Ito, with mosaicism, 53 Hypotriploidy, 289 prenatal diagnosis, 465 H19 gene, 360 ICF syndrome, 346 Ideograms of chromosomes, 501 Imbalance, assessment of, 47 Immunodeficiency, centromere instability, facies syndrome, 346 Immunosuppressive therapy, 494 Implantation failure, 382 Imprinting , 56, 353 erasure, 46 relaxation, 46, 357 Imprinting center, 15q11q13, 362, 365 Inactivation of X chromosome, 112, 221 X-autosome translocation, 115 Industrial agents, risks for offspring , 496 Infertility, 388, 397 FSHR mutation, 390 ICSI, normal chromosomes, 399 Infertility, female, 390, 398 inversion X , 171 reciprocal translocation, 96 rob translocation, sex chromosomes, 390 Turner syndrome, 224 X-X translocation, 135 Infertility, male, 390, 398 AZF deletion, 391 complex rearrangement, 219 IVF, 398 insertion, 189 inversion, 169 reciprocal translocation, 95, 392 ring , 202
INDEX
•
623
Infertility, male (Continued) rob translocation, 148, 392 sex vesicle, 129 supernumerary marker, 239 X aneuploidy, 391 X-autosome translocation, 116 XXXY, 229 XXY, 223 XXYY, 229 X/XY, X/XYY, 228 Y abnormality, 391 Y-autosome translocation, 129, 138 Yq deletion, 391, 399 Insertions, 183 Insertion, interchromosomal, 184 direct and inverted, 183 NOR insertion, 189 prenatal diagnosis, 471 quadrivalent formation, 185 risks to carrier, 191 segregation, 184, 186 sperm studies, 189 two-way, 189 viable recombinant forms, 186 Insertion, intrachromosomal, 194 between-arm, 195 direct and inverted, 194 meiosis, complete synapsis, 196 incomplete synapsis, 195 risks to carrier, 199 within-arm, 194 Instability syndromes, 341 Interchange monosomy, 89 X-autosome translocation, 128 Interchange trisomy, 71 ‘correction’, with UPD, 355 X-autosome translocation, 128 Interchromosomal effect inversion, paracentric, 181 inversion, pericentric, 171 reciprocal translocation, 111 rob translocation, 153 Interchromosomal insertion See Insertion, interchromosomal Intermediate zone fragile X allele, 253 Intrachromosomal insertion See Insertion, intrachromosomal Intrauterine growth retardation, UPD, 357 Inv dup(22) See Isodicentric 22 Inversion, 161 acrocentric, 163 cryptic, 161 frequency, 164 inv(9)(p11q12), 258 locus disruption/deletion, 162 loop formation at meiosis, 164 normal variant, 163 paracentric See Inversion, paracentric pericentric See Inversion, pericentric Inversion, paracentric, 176 confusion with intrachromosomal insertion, 181 dicentric and acentric formation, 176 frequency, 164 innocuousness of most, 62, 180 interchromosomal effect, 181 inv dup from parental paracentric, 178 inv(8)(p23), 180 oöcyte study, 177 position effect, 179
624
•
INDEX
prenatal diagnosis, 182, 471 risks to carrier, 181 sperm studies, 176 U-loop formation, 177 viable recombinant forms, 177 X chromosome, 180 prenatal diagnosis, 471 Y chromosome, 180 Inversion, pericentric, autosomal, 164 cryptic, 161 heterosynapsis, 165 homosynapsis, 164 infertility, 169 innocuousness of some, 169 interchromosomal effect, 169 inv(2)(p11.2q13), 163 meiotic loop formation, 164 normal variant forms, 163, 174 prenatal diagnosis, 471 recombinant forms, 164 risks to carrier, 174 sperm studies, 165 viability of recombinant forms, 166, 167 Inversion, pericentric, X chromosome, 171 gonadal function in female carrier, 171, 175 prenatal diagnosis, 471 risks to female carrier, 171, 175 risks to male carrier, 173, 176 Inversion, pericentric, Y chromosome, 173, 485 risks to carrier, 176 Inv dup 15 See Isodicentric 15 In vitro fertilization See IVF ISCN, 507 ideograms, 501 Isochromosome, 305 complementary, 157, 159 generation, 305 i(Xq), 178 recurrence, 305 risks to parents of (i) child, 332 Isochromosomes of individual chromosomes: i(5p), 305 prenatal diagnosis, 475 i(8p), 305 prenatal diagnosis, 475 i(9p), 305 prenatal diagnosis, 475 i(10p), 305 prenatal diagnosis, 475 i(12p), 305 prenatal diagnosis, 475 i(13q), 154 prenatal diagnosis, 475 i(14q), 154 i(15q), 154 idic(15), 306 prenatal diagnosis, 474 i(18p), 305 gonadal mosaicism, 306 prenatal diagnosis, 476 i(18q), prenatal diagnosis, 476 i(20q), 305 prenatal diagnosis, 476 i(21q), 154, 283 prenatal diagnosis, 476 gonadal mosaicism, 283 recurrence risk, 292 i(22q), 154, 305
prenatal diagnosis, 476 idic(22), 307 prenatal diagnosis, 474 i(Xp), prenatal diagnosis, 484 i(Xq), 305 prenatal diagnosis, 484 i(Y), prenatal diagnosis, 484 Isodicentric Y with disorder of sex development, 336 prenatal diagnosis, 484 two copies, 485 Isodicentric 15, 306 prenatal diagnosis, 474 Isodicentric 22, 307 prenatal diagnosis, 474 Isodisomy, 56, 352 Isozygosity for recessive gene, UPD, 43, 352, 357 rob translocation, 152 IVF abnormality risk, 393 Angelman syndrome, 372 Beckwith-Wiedemann syndrome, 369 epigenetics, 394 Klinefelter syndrome, 224 maternal serum screening , 413 male infertility, 398 multiple pregnancy, 399 prenatal diagnosis, 399 Turner syndrome, 231 Jacobsen syndrome, 319 Jumping translocation, 159, 160 Karyotype, definition, 5 karyotype-phenotype correlations, 48 Kidney cancer, no. 3 translocation, 111 malformations, 48 Kleefstra syndrome, 318 Klinefelter syndrome, 285 artificial reproductive technology, 224 infertility, 223 long-term follow-up, 478 maternal age effect, 407 meiotic origin, 35 mosaicism, 224 natural paternity, 223 partial, X-autosome translocation, 121 prenatal diagnosis, 478 risks to parent of XXY child, 293 risks to XXY man, 230 from ICSI conception, 224 sperm studies, 223 fathers of XXY, 379 Kouska’s fallacy, 63 Langer-Giedion syndrome, 317 Large-headed sperm, 392 Leri-Weill dyschondrosteosis, t(X;Y), 133 Leukemia, reproductive effects, 493 Lines of Blaschko, 53 Lissencephaly, 322 Locus disruption, 162, 308 Loss of sex chromosome with ageing , 32, 226 Low-copy repeats, 296 Low-level X/XX mosaicism, 225, 231 Lyonization, 112
Male infertility See Infertility, male Marker chromosome See Supernumerary marker chromosome Massively parallel sequencing , 25 prenatal diagnosis, 424 Maternal age aneuploidy (other than DS) risks, 407 cutoff for ‘advanced’, 404 Down syndrome, 403, 405 ethnic comparisons, 406 hydatidiform mole, 394 meiotic apparatus decline, 39 miscarriage risk, 387 nondisjunction association, 403 oöcyte abnormality, 39, 378 preimplantation diagnosis, 427 Prader-Willi syndrome, 364 recurrent miscarriage, 397 risk tables, 406–409 secular changes, 407 Silver-Russell syndrome, 359 UPD, 353 Maternal blood, fetal cells from, 423 Maternal hypomethylation syndrome, 394 Maternal serum screening for fetal aneuploidy, 410 ethical issues, 415 false-positive result, previous pregnancy, 416 first trimester, 411 free fetal DNA , 411, 423 information-giving , 414 interpretation, 414 IVF pregnancy, 413 prevalence of DS, effect upon, 415 rare trisomies, 416 second trimester, 412 triploidy, 416 twin pregnancy, 413 Meiosis, 28 Meiosis I recombination, 71 Meiosis II nondisjunction, 93 Meiotic drive, 94, 145 Mendel’s second law, 28 Mental retardation, 9 chromosomal causes, 10 pregnancy and sterilization, 17 Methylation FRAXA , 245 Beckwith-Wiedemann syndrome, 361 Prader-Willi syndrome, 370 Angelman syndrome, 370 Silver-Russell syndrome, 361 Miconazole, mouse sperm study, 494 Microarray analysis, 22 blastocyst biopsy, 428 copy number variants, 269 ethical issues, 16 ‘first tier’ test, 22 FISH confirmation, 270 incidental discoveries, 271 interpretation, 270, 272, 273 methodology, 25 preimplantation diagnosis, 435 precision of, 51 prenatal diagnosis, 274 1q21.1 deletion, 273 16p11.2 deletion, 273 Microdeletion See Deletion Microduplication X , 229 Microöphthalmia and linear skin defects, 134
INDEX
•
625
Miller-Dieker syndrome, 322 Minute supernumerary chromosome, minSMC, 304 See also Supernumerary marker chromosome Miscarriage, 377, 384 counseling , 396 jumping translocation, 159 karyotyping products, 384 maternal age, 397 preimplantation diagnosis, 429 recurrent miscarriage, 386, 396 due to rearrangement, 387 risks to couple, 396 Turner mosaic, 231 Mismatch repair genes, aneuploidy, 57 Mitochondrial mutations, nondisjunction, 39 Mole See Hydatidiform mole Monosomic rescue See Correction of monosomy Monosomy, autosomal, 285 Monosomy X See Turner syndrome Morula, 381 Mosaic trisomy at prenatal diagnosis See specific karyotype Mosaic variegated aneuploidy, 347 Mosaicism, 31, 52 amniotic fluid, 56, 453 chaotic See Chaotic mosaicism confined placental 56, 442, 449, 456 constitutional, definition, 7 Down syndrome, 284 embryo, 32, 435 full aneuploidy, 52 generation, 31 gonadal See Gonadal mosaicism hypomelanosis of Ito, 53 placental See Confined placental mosaicism prenatal diagnosis See Prenatal diagnosis, mosaicism ring chromosome, 202 somatic-gonadal See Somatic-gonadal mosaicism structural rearrangement, 53 tissue sampling , 54 translocation, balanced, 53 translocation, unbalanced, 53 trisomy at PND See individual Trisomy entries variegated aneuploidy syndrome, 347 X ‘normal mosaicism’, 225 Multiplex amplifiable probe hybridization, 25 Neocentromere discovery, 11 ring chromosome, 209 SMC, 307 Neuropathy Charcot-Marie-Tooth, 330 pressure palsies, 323 Next-generation sequencing , 25 prenatal diagnosis, 424 Nijmegen breakage syndrome, 345 NLRP7 gene, 395, 400 Nomenclature, 4, 507 Nonallelic homologous recombination, 296 Nonconjugation, 37 Nondirective counseling , 13 professional differences, 441 Nondisjunction, 34 causes, 39 predisposition to, 57 two-hit hypothesis, 277 Nonhomologous end-joining , 296 Nonhomologous rob See Robertsonian translocation, heterologous
626
•
INDEX
Non-Robertsonian dicentric See Telomeric fusion NOR , 260 interstitial insertion, 262 prenatal diagnosis, 477 robertsonian translocation, 143 staining , 22 translocation of, 262 variation, 260 Normal variants, 257, 264 prenatal diagnosis, 476 Noxious agents, chromosomal damage, 489 Nuchal translucency, 411, 418 Nucleolar organizing region See NOR Nulliallelic expression, 353 Occult abortion, 382 Oligospermia, 390 definitions, 391 reciprocal translocation, 95 rob translocation, 148 Y-autosome, 130 Oöcyte donation, Turner syndrome, 231 giant binucleate, 379 karyotyping , 72, 378 Opposite imbalances See Complementary rearrangements Ovarian failure See Premature ovarian failure Ovarian mosaicism See Gonadal mosaicism Ovarian teratoma, 368 Ovotesticular disorder of sex development, 336 counseling , 340 familial, 337 Ovum See Oöcyte Packaging of chromosomes, 7 Pallister-Killian syndrome, 305 PAR See Pseudoautosomal region Paracentric inversion See Inversion, paracentric Paralogous sequence See duplicon Parental age, aneuploidy risk, 403 Partial hydatidiform mole See Hydatidiform mole Patau syndrome See Trisomy 13 Paternal age, 404 PCR-based methodologies, 24 Pelizaeus-Merzbacher disease, 331 Percutaneous umbilical blood sampling See Fetal blood sampling Perfect pitch, 97 Pericentric inversion See Inversion, pericentric Perinatal death, chromosomal causes, 8, 10 Periventricular nodular heterotopia, as epiphenomenon, 48 Pesticide exposure, sperm studies, 496 Phelan-McDermid syndrome, 327 Phenotype-first approach, 11 Phenotypes due to imbalance, 8 Pigmentary anomalies with mosaicism, 53 Placental biopsy, 423 Placental dysfunction, 452 Placental mesenchymal dysplasia, 396, 400 Placental mosaicism, confined, 56, 442, 449, 456 placental effects, 452 Pleural effusion sampling, fetal, 424 Polar bodies, 28 biopsy, 430 karyotyping , 378 oöcyte nondisjunction, 378 preimplantation diagnosis, 430 Polymorphic chromosomes, 257 Polyploidy, 286
Polyposis adenomatous, deletion 5q, 315 juvenile, deletion 10q, 315 Polysomy sex chromosomes, 286 infertility, 232 meiotic origin, 36 prenatal diagnosis, 480 risk of recurrence, 286 Position effect, 45 inversion, 179 reciprocal translocation, 96, 109 Potocki-Lupski syndrome, 331 Potocki-Shaffer syndrome, 195, 318 Prader-Willi syndrome, 362 critical region, 56 deletion, 362 due to rearrangement, 363 with Y;15 translocation, 133, 363 imprinting center defects, 362, 365 maternal age effect, 364 phenocopy, 371 risks to parents of PWS child, 370 with trisomy 15 mosaicism, 364 UPD, 364 Precocious division, as cause of nondisjunction, 37 Predictive testing , 15 Predisposition to aneuploidy, 57 Predivision, as cause of nondisjunction, 37 Pre-embryo, 380 Preimplantation diagnosis (PGD), 426 aneuploidy screening , 427 controversy, 428 recurrent miscarriage, 429 blastocyst biopsy, 430 blastomere biopsy, 430 carriers of rearrangements, 427 clinical procedures, 429 counseling , 436 gender selection, 429 implantation failure, 429 ‘IVFlings’, 438 laboratory procedures, 429, 432, 436 microarray, 435 mosaicism, chaotic, 435 polar body biopsy, 430 reciprocal translocation, 110 recurrent miscarriage, 429 risks of imbalance, 437 Premature centromere separation Roberts syndrome, 345 variegated aneuploidy, 347 Premature menopause See Premature ovarian failure Premature ovarian failure fragile X premutation, 248, 253 with translocation, 96 Turner variant, 231 X;autosome translocation, 116 Premutation, fragile X , 243 neurological syndrome in males, 247 premature ovarian failure, 248 premutation, 243 ‘high-end’, ‘low-end’, 253 Prenatal diagnosis, 439 (Specific karyotypes are listed individually) access to, 18 amniocentesis, 422 culture failure, 423 early, 423 mosaicism, 453
anxiety associated, 425 apparently balanced rearrangement, 466 applied embryology, 442 carrier state, 467 celocentesis, 424 cervical lavage, 424 complex rearrangement, 468 confined placental mosaicism, 442, 449 placental effects, 452 copy number variants, 441 cordocentesis, 423 counseling, differing approaches, 440 chorionic villus sampling , 422 direct cf. long-term, 422 false-negative results, 450 limb defect risk, 422 sampling , 452 trisomies, interpretation, 451 UPD, 452 cystic hygroma sampling , 424 decision-making , 439 de novo apparently balanced rearrangement, 466 complex , 468, 469 insertion, 471 inversion, 469, 471 mosaicism, 468 reciprocal translocation, 467 ring, autosomal, 471 risks, 466 rob translocation, 469, 470 whole-arm translocation, 468, 469 X-autosome translocation, 469 Y-autosome translocation, 469 de novo unbalanced rearrangement, 471 X-autosome translocation, 472 Y-autosome translocation, 472 diploid/tetraploid mosaicism, 466 diploid/triploid mosaicism, 465 embryology, 442 fetal blood sampling (cordocentesis), 423 fetal cells from maternal blood, 423 FISH, 420 insertion, de novo, 471 inversion, de novo, 471 isochromosomes, 474 iso(5p), 475 iso(8p), 475 iso(9p), 475 iso(10p), 475 iso(12p), 475 iso(13q), 475 iso(18p), 476 iso(18q), 476 iso(20q), 476 iso(21q), 476 iso(22q), 476 maternal serum screening , 410 microarray, 274, 421, 441 MLPA , 421 mosaicism, 442 amniocentesis, 453 complex rearrangement, 469 confined placental, 442, 449 CVS, 450, 456 ‘ICF-like’ chromosomes, 476 isochromosome, 474 laboratory assessment, 446 levels, 446, 450, 453
INDEX
•
627
Prenatal diagnosis (Continued) mechanisms, 444, 448 normal variants, 476 reciprocal translocation, 468 rob translocation, 469 sex chromosome, 477 structural rearrangement, 471 trisomies See Trisomies UPD risk, 452 whole-arm translocation, 469 next-generation sequencing , 424 non-invasive, 423 nuchal translucency, 418 painful, 425 phenotype prediction, 448 pleural effusion sampling , 424 polyploidy, 465 proteomic fingerprinting , 424 QF-PCR , 421 reciprocal translocation, 466, 467 mosaicism, 468 residual (occult) low-level trisomy, 456, 470 ring autosomal, 476 X , 484 Y, 485 rob de novo balanced heterologous, 470 homologous, 470 sex chromosome abnormality decision-making , 440 predicted phenotypes, 477 mosaicisms, 481 polysomies, 480 structural rearrangement, 466 de novo apparently balanced, 466 unbalanced, 471 supernumerary chromosome, 472 bisatellited, 474 isochromosome see above isodicentric 15, 474 isodicentric 22, 474 ring , 476 tetraploidy, 465 mosaicism, 466 triploidy, 465 mosaicism, 465 trisomy 21, decision-making , 440 trisomy mosaicism See individual trisomies twin pregnancy, 419 discordant karyotypes, 454 UPD rob, 470 trisomy 15, 459, 463 trisomy 16, 459, 463 ultrasound anomalies, 417 whole-arm translocation, de novo, 468 X-autosome translocation balanced de novo, 469 familial, 137 unbalanced, 472 X deletion, 483 X duplication, 483 X ring , 484 X and Y mosaicisms, 482 X and Y polysomies, 480
628
•
INDEX
XX male, 481 XXX , 478 X/XX , 482 XX/XY, 481 XXY, 478 X-Y translocation, 483 XYY, 479 Y abnormality, 484 isochromosome, 484 ring , 485 Y-autosome translocation de novo balanced, 469 unbalanced, 472 45,X , 479 Pressure-sensitive palsy, 323 Prevalence figures cf. maternal age (Tables) trisomy 13, 408 trisomy 18, 408 trisomy 21, 406, 407 XXX , 408 XXY, 408 Prevention, primary, 293 Previous pregnancy, increased-risk screen, 416 Probability, 59 Products of conception, 384 Proteomic fingerprinting, PND, 424 Pseudoautosomal regions, 113, 129 Pseudohypoparathyroidism, UPD 20, 367 Psychosexual issues Turner syndrome, 480 XY female, 339 Pure gonadal dysgenesis, XY, 334 Pyrimethamine, mouse sperm study, 494 Q-banding , 21 Quadrivalent, 69 Qualitative assessment of imbalance, 47 Quantitative assessment of imbalance, 47 R-banding , 21 Radioactivity, 495 Radioisotopes, Radiotherapy See Cancer Radiology, risk to child, 494 Rearrangement formation nonallelic homologous recombination, 296 nonhomologous end-joining , 296 Reciprocal translocation, autosomal, 67 assisted reproduction, 96 balanced translocation in fetus, 109 cancer association, 111 carrier couple, 97 counseling , 99 de novo apparently balanced, 95 embryo, segregations, 75 frequency, 68 infertility, 95, 389 interchromosomal effect, 111 locus disruption, 96 microarray, 101 miscarriage, 110 mosaicism, 98 preimplantation diagnosis, 110 position effect, 96 risks to carrier, 99, 103 preimplantation diagnosis, 437 prenatal diagnosis, 101
segregation modes, 69, 77 single and double segment, 68 t(4;8)(p16;p23), 80 t(11;22)(q23;q11), 86, 107 unstable, 98 UPD, 107 whole arm, 69, 80 Reciprocal translocation, X-autosomal See X-autosome translocation Recombination, somatic, 34 Recurrence risks, 59 Recurrent abnormalities, 57 Recurrent miscarriage See Miscarriage Reifenstein syndrome, 339 Relaxation of imprinting , 46, 357 Replication banding , 22 Research applications, 11 participation in, 19 Rescue of monosomy See Correction of monosomy Rescue of trisomy See Correction of trisomy Residual low-level trisomy prenatal diagnosis, 452 rob, 148 Retinoblastoma, with 13q deletion, 319 Reverse banding , 21 Ring chromosome, 201 balancing deletion, 209, 210 café-au-lait macules, 207 epilepsy, ring 20, 206 formation, 202 general ring syndrome, 202 individual types See next heading infertility, male, 202 multiple rings, 210 mosaicism, dynamic, 202 neocentromere, 209 neurofibromatosis type 2, ring 22, 207 prenatal diagnosis, 476 risks to carrier karyotype 46(r), 210 karyotype 47,+(r), 210 mosaic 46,N/46(r), 202 segregation, 202 supernumerary small ring , 207 Rings of individual chromosomes: ring 1, 202 ring 2, 202 ring 4, 204 ring 6, 204 ring 7, 204 ring 8, 204 ring 9, 204 ring 10, 204 ring 11, 204 ring 12, 204 ring 13, 205 ring 14, 205 ring 15, 205 ring 17, 205 ring 18, 206 ring 19, 206 ring 20, 206 ring 21, 206 ring 22, 207 ring X , 227 tiny, 484 ring Y, prenatal diagnosis, 485
supernumerary ring 1, 207 supernumerary ring 2, 208 supernumerary ring 3, 208 supernumerary ring 4, 208 supernumerary ring 7, 208 supernumerary ring 8, 208 supernumerary ring 10, 208 supernumerary ring 12, 209 supernumerary ring 15, 209 supernumerary ring 18, 209 supernumerary ring 20, 209 supernumerary ring 22, 209 tiny ring X,prenatal diagnosis, 484 Risk and risk figures, 59, 64 confidence limits, 64, 510 private risk figure, 61 Riyadh chromosome breakage syndrome, 341 Roberts syndrome, 345 Robertsonian fission, 149, 159, 160 Robertsonian translocation, heterologous 140 couple both carriers, 149 dicentric, 143 formation, 142 frequency, 142 infertility, 148, 150 interchromosomal effect, 153 isochromosome, 143, 149 isozygosity for recessive gene, 152 meiotic drive, 145 miscarriage, 150 monosomic correction, 148 mosaicism, 149 NORs, 143 oöcyte karyotyping , 146 preimplantation diagnosis, 153 prenatal diagnosis, de novo balanced, 470 UPD, 152 recurrent miscarriage, 148, 150 risks to carrier, 151 13q14q, 151 13q15q, 151 13q21q, 152 13q22q, 152 14q15q, 152 14q21q, 151 14q22q, 152 15q21q, 152 15q22q, 152 21q22q, 152 UPD, 145, 152 segregation, 144, 146 sperm karyotyping , 146 translocation DS de novo, 284 familial, 151, 152 trisomic correction, 145 UPD, 152 Robertsonian translocation, homologous, 149 monosomic rescue, 150 prenatal diagnosis, 153, 470, 476 risks to carrier, 153 segregation, 149 translocation DS de novo, 283, 476 familial, 154 trisomic correction, 150
INDEX
•
629
Rubinstein-Taybi syndrome, 321 Russell-Silver syndrome See Silver-Russell syndrome Saethre-Chotzen syndrome, 316 Satellite DNA , 6 Satellites, acrocentric chromosomes, 259 translocation of, 262 Satellite, interstitial insertion, 263 prenatal diagnosis, 477 Screening See Maternal serum screening for fetal trisomy Seckel syndrome, 347 Segmental aneusomy, 46, 296 Segmental UPD, 356 Segregation, 29 analysis, 60 autosomal translocations, 69 adjacent-1, 71, 78 adjacent-2, 71, 82 alternate, 71 interchange monosomy, 89 interchange trisomy, 89 more than one type, 90 tertiary monosomy, 87 tertiary trisomy, 86 3:1, 71, 86 4:0, 71, 89 insertions, 184, 186 inversions, 164 X-autosome translocations, 121 adjacent-1, 124 adjacent-2, 124 interchange trisomy, 128 tertiary monosomy, 126 4:0, 128 Seminoma, 492, 493 Sex chromosome imbalance, 52 prenatal diagnosis, 477 Sex chromosome polysomy See Polysomy sex chromosomes Sex chromosome, structural abnormality prenatal diagnosis, 483, 484 Sex chromosome vesicle and infertility autosomal translocation, 95 Y-autosome translocation, 129 Sex hormone treatment, 494 Sex reversal See also XX male, XY female campomelic dysplasia, 338 del 9p24.3, 317 Sex vesicle, 129 Shprintzen syndrome, 326 Silver-Russell syndrome, 360, 369 epimutation, 369 phenocopy, 370 UPD 7, 360 Silver (Ag) staining , 22, 260 Single nucleotide polymorphism ethical issues, 26 microarray, 24 preimplantation diagnosis, 428, 435 prenatal diagnosis, 448 Single segment exchange autosomal translocation, 68 X-autosome translocation, 121 Sister chromatid exchange Bloom syndrome, 344 Skewing of X-inactivation, 113 Small supernumerary chromosome, sSMC, 304 See Supernumerary marker chromosome Smith-Magenis syndrome, 323
630
•
INDEX
generation of deletion, 296 SNP See Single nucleotide polymorphism Solid staining , 21 Somatic-gonadal mosaicism, 54 Somatic recombination, 34 segmental UPD, 356 Sotos syndrome, 315 Sperm aneuploidy rate, 379 banking , 493 caffeine drinking , 496 cancer therapy, 490 cigarette smoking , 496 defects, 392 donor, abnormal child, 399 fathers of aneuploid children, 379 Down syndrome children, 282 Klinefelter syndrome boys, 286 Turner syndrome girls, 286 gonadal mosaicism, 54 Hodgkin’s lymphoma, 492 immunosuppressive therapy, 494 infertility, 393 insertion, interchromosomal, 189 inversion, pericentric, 165 karyotyping , 379 Klinefelter syndrome, 223 ‘large-headed’, 392 ‘packaging’, 380 radiotherapy, 492 reciprocal translocation carrier, 72 rob translocation, 144 seminoma, 492, 493 ‘tail stump syndrome’, 392 testicular cancer, 493 trisomy 8 mosaicism, 234 trisomy 18 mosaicism, 234 X aneuploidy, 391 XXY, 223 X/XY, 228 X/XYY, 483 X-Y translocation, 134 XYY, 224 Y-autosome translocation, 130 Spontaneous abortion See Miscarriage SRY gene, 334 hermaphroditism, 337, 340 ovotesticular disorder, 337, 340 paternal gonadal mosaicism, 334 XX male, 134, 335 XY female, 334 45,X male, 134, 336 Stable non-Robertsonian dicentric See Telomeric fusion Standard error, measurement of, 510 Starburst multiradial, ICF syndrome, 347 Sterilization in the mentally retarded, 18 Streak gonadal dysgenesis, 335 Structural rearrangement, 44 prenatal diagnosis, 466 Subtelomeric rearrangements, 22, 82, 106 risks to carrier, 106 polymorphisms, 266 Supernumerary marker chromosome (SMC), 239, 304 balancing , 159 familial transmission, 239 harmless SMC, 241 infertility in male, 239
inv dup(15) See Isodicentric 15 mosaicism, 240 neocentromere, 307 parent a carrier, prenatal diagnosis, 472 chromosome 10-derived, 474 chromosome 18-derived, 421 de novo, 473 familial, 473 idic(15), 474 idic(22), 474 isochromosomes, 305, 474 ring , 476 ring See Ring chromosomes risks to carrier, 239 risks to parent of SMC child, 332 segregation, 240 UPD, 356 Swyer syndrome, 334, 338 Synapsis, in meiosis, 29 heterosynapsis, 30 homosynapsis, 30 Synaptonemal complex , 29 Syngamy, 28 SYPC3 gene, 39, 57 Tandem duplication, 303 TAR See Thrombocytopenia-absent radius Telomere, 6, 229 Telomeric fusion translocation, 158, 159 Teratoma, ovarian, 57, 368 Termination of pregnancy, decision-making , 439 Tertiary monosomy autosomal, 87, 107 X-autosomal, 126 Tertiary trisomy autosomal, 71, 86 Testicular feminization See Androgen insensitivity Testicular mosaicism See Gonadal mosaicism Testicular sperm extraction, 224 Tetraploidy, 289 diploid/tetraploid mosaicism, 289 prenatal diagnosis, 466 prenatal diagnosis, 465 risks to parent, 293 Tetrasomy, 36 Therapy Fragile X , 254 del(15)(q13), 320 Three-way translocation, 213 Thrombocytopenia-absent radius, 273 Tiny ring X syndrome, prenatal diagnosis, 484 Tobacco use, risks to offspring , 496 Tomaculous neuropathy, 323 Transient neonatal diabetes, 359 Translocation See Apparently balanced rearrangement Complex chromosome rearrangement Embryo, translocation carriers Insertion Jumping translocation Reciprocal translocation Robertsonian translocation Telomeric fusion translocation Unstable familial translocation Whole arm translocation X-autosome translocation
11;22 translocation Translocation (rob) DS, 140, 144, 149, 151, 154 Translocation (rob) trisomy 13, 144, 151 Translocation santeuse See Jumping translocation Translocation t(11;22)(q23;q11) See 11;22 translocation Tremor-ataxia syndrome, fragile X , 247, 248 Tricho-rhino-phalangeal syndrome, 317 Trinucleotide repeat expansion, 46, 243, 244 Triplet repeat See Trinucleotide repeat Triplication, 238, 304, 332 Triploidy, 286 abortion, 288 prenatal diagnosis, 465 diandry, 287 digyny, 288 diploid/triploid mosaicism, 289 frequency, 396 mosaicism, at PND, 465 phenotype, 288 risks to parent, 293 Trisomic rescue See Correction of trisomy Trisomy abortion, 384 double, 36 maternal age association, 403 meiotic origin, 34, mosaicism See individual Trisomy entries residual See Residual low-level trisomy Trisomy for specific chromosomes: Trisomy 1, 382 Trisomy 2 acardius, 34 mosaicism detection at amniocentesis, 460 detection at CVS, 456 Trisomy 3 acardius, 386 mosaicism detection at amniocentesis, 461 detection at CVS, 457 Trisomy 4 mosaicism detection at amniocentesis, 461 detection at CVS, 457 Trisomy 5 mosaicism detection at amniocentesis, 461 detection at CVS, 457 Trisomy 6 mosaicism detection at amniocentesis, 461 detection at CVS, 457 Trisomy 7, 285 mosaicism detection at amniocentesis, 461 detection at CVS, 457 phenotypes, 461 placental, 451 Trisomy 8 , 285 embryo, 382 mosaicism cancer risk, 241 cause, 41 detection at amniocentesis, 461 detection at CVS, 458 risks to carrier, 233, 240 sperm studies, 234
INDEX
•
631
Trisomy 9, 285 mosaicism detection at amniocentesis, 462 detection at CVS, 458 prenatal diagnosis, 455 survival to term, 285, 458 Trisomy 10, 285, 455 mosaicism detection at amniocentesis, 462 detection at CVS, 458 Trisomy 11 mosaicism detection at amniocentesis, 462 Trisomy 12 mosaicism detection at amniocentesis, 462 detection at CVS, 458 Trisomy 13, Patau syndrome, 285 maternal age association, 285 meiotic origin, 285 mosaicism detection at amniocentesis, 454, 462 detection at CVS, 458 very low-level, 453 prenatal diagnosis, 454 recurrence, 293 spontaneous abortion, 404 survival, 454 Trisomy 14, 285 mosaicism detection at amniocentesis, 462 detection at CVS, 458 survival to term, 285, 455 Trisomy 15 mosaicism detection at amniocentesis, 463 detection at CVS, 459 Prader-Willi syndrome, 364 Trisomy 16 ‘correction’ with UPD, 357, 367, 450 maternal age association, 404 meiotic origin, 34, 450 miscarriage, 384 mosaicism detection at amniocentesis, 463 detection at CVS, 367, 459 eclampsia risk, 463 mild phenotype, 53 16p, 124 Trisomy 17 double, 36 miscarriage, 382 mosaicism detection at amniocentesis, 463 Trisomy 18, Edwards syndrome, 285 clinical management, 400, 454 maternal screening , 416, 418 meiotic origin, 285 mosaicism detection at amniocentesis, 463 detection at CVS, 459 recorded cases in adults, 234 parental mosaicism, 234 perinatal death, 386 prenatal diagnosis, 454 false positive, 421 recurrence, 285, 293 spontaneous abortion, 404
632
•
INDEX
survival, 404, 454 Trisomy 19 mosaicism detection at amniocentesis, 464 Trisomy 20 maternal, 211 mosaicism detection at amniocentesis, 464 detection at CVS, 459 prenatal diagnosis, 455 Trisomy 21, Down syndrome alcohol, maternal, 496 Alzheimer association, 280 caffeine, maternal, 496 cigarette smoking, maternal, 496 critical region, 278 cytogenetic forms, 280, 284 de novo rob DS, 284, 292 DSCR1, 278 epidemiology, 407 familial rob DS, 151, 152, 154, 284 family history DS, 283 fathers of DS, sperm study, 379 genotype-phenotype, 278 gonadal mosaicism, parental standard trisomy, 54, 282 i(21q), 283 heterotrisomy, 293 interchromosomal effect, 284 isochromosome 21q, 283 maternal age association, 291, 403 maternal serum screening , 410 meiotic origins, 280 molecular biology, 278 mosaicism, 283 detection at amniocentesis, 464 detection at CVS, 460 generation, 283 parental, 235, 282 risks to carrier, 241 risks to parent of mosaic child, 283, 290 oögenesis, 234 parent with DS, 234, 241 phenotypic map, 279 prenatal diagnosis, 455 prevalence, 409 effect of elective abortion, 410 effect of population screening , 410 secular changes, 407 rare chromosomal causes, 284 rob(21q21q) See Isochromosome, i(21q) rob translocation DS, de novo, 284 familial, 151, 152, 284 risks to parent of child with de novo rob DS, 284 familial rob DS, 151, 152, 284 family history DS, 283, 292 i(21q), 292 mosaic standard trisomy, 282 products of conception +21, 292 reciprocal translocation, 284 standard trisomy, 283, 290 sperm study in fathers, 379 spermatogenesis, 235 spontaneous abortion, 404 standard trisomy, 280 Trisomy 22
maternal serum screening , 416 mosaicism, 53 detection at amniocentesis, 465 detection at CVS, 460 prenatal diagnosis, 455 True hermaphroditism See Ovotesticular disorder sex development Tuberous sclerosis, 16p translocation, 89 Turner syndrome, 224, 286 clinical phenotype, 479 deletion X , 226 due to X-autosome translocation, 117 premature ovarian failure, 227 risks to carrier, 226 prenatal diagnosis, 479 imprinting of parental X , 368 IVF, 231 infertility, 224 i(Xq), 226 long-term follow-up, 480 meiotic origin 45,X , 379 mosaicism, 225 risks to mosaic TS woman, 231 oöcyte loss, 224 ovum donation, 231 phenotype, parental origin of X , 368 predicted at PND, 480 recurrence risk, 286, 293 ring X , 227 risks to mosaic TS woman, 231 risks to parent of TS child, 285, 293 risks to TS woman, 230 sperm studies in fathers, 379 tiny ring X , 484 variant forms, 225, 226 risks to carrier, 231 X-autosome translocation, 117 Twins, 33 discordant karyotype, 33, 42 prenatal diagnosis, 419 vanishing , 386 UBE3A gene, Angelman syndrome, 362, 366 UKCAD, 309 Ullrich-Turner syndrome See Turner syndrome Ultrasonography for fetal defect, 417 screening , 411 Uniparental diploidy, 394 Uniparental disomy (UPD), 46, 56, 351 Angelman syndrome See Angelman syndrome Beckwith-Wiedemann syndrome, 360, 369 complete, 57, 368 mosaic, 368 ovarian teratoma, 368 correction of monosomy, trisomy, 42, 145, 148, 353 double, 368 gametic complementation, 37, 353 generation, 42, 353 heterodisomy, 352 isochromosome formation, 356 isodisomy and isozygosity 44, 352, 358 intrauterine growth retardation, 357 maternal age association, 353 miscarriage, 358 monosomic rescue, 353 Prader-Willi syndrome See Prader-Willi syndromes prenatal diagnosis, 374
risks to parent of UPD child, 368 rob translocations, 145, 148 segmental, 57, 356 Silver-Russell syndrome, 360, 369 supernumerary marker, 356 transient neonatal diabetes, 359 trisomic rescue, 353 trisomy, residual mosaic, 357 Uniparental disomy for specific chromosomes: UPD 1, 358 UPD 2, 358 UPD 3, 358 UPD 4, 359 UPD 5, 359 UPD 6, 359 UPD 7, 359 UPD 8, 359 UPD 9, 359 UPD 10, 359 UPD 11, 359 UPD 13, 361 UPD 14, 361 UPD 15, 362 UPD 16, 367 UPD 17, 367 UPD 20, 367 UPD 21, 367 UPD 22, 367 UPD X , 367 Unmasking of recessive gene, 300 VACTERL syndrome, 342 Vanishing twin, 386 Variant chromosomes, 257 Variegated aneuploidy, 346 Viability of imbalances translocations, 100, 102 WAGR syndrome, 318 Warsaw breakage syndrome, 347 Whole arm translocation, 69, 80 prenatal diagnosis, 468 mosaicism, 469 Williams syndrome, 316 Williams-Beuren syndrome, 316 Wilms tumor Beckwith-Wiedemann syndrome, 361 survivors’ children, 592 WAGR syndrome, 318 Wolf-Hirschhorn syndrome, 312 translocation, 168 Wrongful handicap, 17 X-autosome translocation, 112 female carrier, 121, 136 inactivation pattern in female carrier, 114 inactivation pattern in unbalanced offspring , 117 infertility, male, 116 locus disruption, de novo translocation, 120 male carrier, 116, 121, 128 prenatal diagnosis balanced de novo, 469 familial, 137 unbalanced, 472 risks to female carrier, 136 risks to male carrier, 137 segregation patterns, 121
INDEX
•
633
X chromosome inactivation, 112 in extra X states, 9 X critical regions, 116 X deletion, 226 microdeletion, 228 prenatal diagnosis, 483 risks to carrier, 231 transmission, 226 X disomy, functional duplication X in male, 229 tiny ring X syndrome, 484 t(Xq;Yq), 135 X-autosome translocation, 115, 117, 119, 124, 126, 135 X duplication, 229 inactivation, unpredictability of, 229 prenatal diagnosis, 483 risks to carrier, 229, 232 Xp11.22-11.23 microduplication, 229 X inactivation, 112, 221, 222 X isochromosome generation, 178 prenatal diagnosis, 484 Xq, 226 X loss with ageing , 226 X microdeletion, 228 risks to carrier, 232 X microduplication, 229 X monosomy See Turner syndrome as cause of miscarriage, 384 X mosaicism, low-level normal, 225 X polysomy See Polysomy sex chromosomes X pseudoautosomal regions, 113 X ring , 226 prenatal diagnosis, 484 tiny ring X syndrome, 484 X skewing of inactivation, 113, 222 XIC, 113 XIST, 113 Xp11.22-p11.23 microduplication, 229 Xp-Yp translocation, 133, 134 Xq-Yq translocation, 135 X-rays, maternal, 494 XX male, 335 counseling , 339 Xp-Yp translocation, 134 XX true hermaphroditism, 336 X-X translocation, 135, 139 XXX female, 285 maternal age effect, 407 meiotic origin, 35 premature ovarian failure, 223 prenatal diagnosis, 478 risks to parent of XXX child, 293 risks to XXX woman, 223, 230 XXX male, 336 X/XX mosaicism, 225, 231 low-level, 225 prenatal diagnosis, 482 XXXX , 229, 232, 286, 480 X/XXX mosaicism, 225 prenatal diagnosis, 482 XXXXX See Polysomy sex chromosomes X/XXX/XX mosaicism, 41, 225 prenatal diagnosis, 482 XXXXY See Polysomy sex chromosomes XXXY, 229, 232, 286 XX/XY chimerism, 337 prenatal diagnosis, 481
634
•
INDEX
X/XXY mosaicism, 228 XXXYY See Polysomy sex chromosomes XXY See Klinefelter syndrome X/XY male, 228 ovotesticular disorder, 337 prenatal diagnosis, 482 XXY/XY mosaicism, 223, prenatal diagnosis, 482 XXYY, 229, 232 prenatal diagnosis, 480 X/XYY, prenatal diagnosis, 483 X/XYY/XY, prenatal diagnosis, 483 XXYYY See Polysomy sex chromosomes XY female, 334 different genetic forms, 334, 335 genetic counseling , 338, 339 management, 339 X-Y translocation classical, 133 microöphthalmia and skin defects, 134 prenatal diagnosis, 483 risks to carrier, 139 sperm analysis, 134 variant forms, 135 Xp-Yp translocation, 134 ‘XYq–’, 135 XYY male, 285 long-term follow-up, 479 origin of extra Y, 36 prenatal diagnosis, 479 risks to parent of XYY child, 293 risks to XYY man, 224, 230 sperm studies, 224 XYYY See Polysomy sex chromosomes XYYYY See Polysomy sex chromosomes Y-autosome translocations, 129, 261 acrocentric p arm-Yqh, 130, 261 de novo Yq;1q, 133 infertility, 129, 138 preimplantation diagnosis, 138 prenatal diagnosis, 469 rare forms, 132 risks to carrier, 130, 138 sperm analysis, 130 45,X male, 132 Y chromosome, 129 loss with ageing , 32 pseudoautosomal regions, 113 Y isochromosome disorder of sex development, 336 infertility, 391 prenatal diagnosis, 484 Y pericentric inversion, 173, 485 Y structural rearrangement prenatal diagnosis, 484 Yq microdeletion, 392 Yq duplication, 229 Yqh translocation acrocentric, 130 nonacrocentric, 132 Yqh variation, 258, 485 Y-ring, prenatal diagnosis, 485 Y-X translocation See X-Y translocation Y-Y translocation, 135 Zygote, 380